Language selection

Search

Patent 2767972 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 2767972
(54) English Title: DUAL VECTOR FOR INHIBITION OF HUMAN IMMUNODEFICIENCY VIRUS
(54) French Title: VECTEUR DOUBLE POUR L'INHIBITION DU VIRUS DE L'IMMUNODEFICIENCE HUMAINE
Status: Deemed Abandoned and Beyond the Period of Reinstatement - Pending Response to Notice of Disregarded Communication
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 15/85 (2006.01)
  • A61K 31/713 (2006.01)
  • A61K 38/16 (2006.01)
  • A61P 31/18 (2006.01)
  • C7K 14/16 (2006.01)
  • C7K 14/47 (2006.01)
  • C7K 14/705 (2006.01)
  • C7K 19/00 (2006.01)
  • C12N 5/10 (2006.01)
  • C12N 15/113 (2010.01)
  • C12N 15/12 (2006.01)
  • C12N 15/62 (2006.01)
  • C12N 15/86 (2006.01)
  • C12N 15/867 (2006.01)
(72) Inventors :
  • CHEN, IRVIN (United States of America)
  • AN, DONG SUNG (United States of America)
  • MILLINGTON, MICHELLE (Australia)
  • BOYD, MAUREEN (Australia)
  • SYMONDS, GEOFFREY P. (Australia)
  • BRETON, LOUIS RANDALL (United States of America)
(73) Owners :
  • THE REGENTS OF THE UNIVERSITY OF CALIFORNIA
  • CSL BEHRING GENE THERAPY, INC.
(71) Applicants :
  • THE REGENTS OF THE UNIVERSITY OF CALIFORNIA (United States of America)
  • CSL BEHRING GENE THERAPY, INC. (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2010-05-26
(87) Open to Public Inspection: 2011-01-20
Examination requested: 2015-05-01
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2010/036247
(87) International Publication Number: US2010036247
(85) National Entry: 2012-01-12

(30) Application Priority Data:
Application No. Country/Territory Date
61/225,687 (United States of America) 2009-07-15
61/287,599 (United States of America) 2009-12-17

Abstracts

English Abstract

The present invention provides an expression vector for preventing or inhibiting HlV entry, fusion or replication in mammalian cells. In particular, the invention provides a recombinant retroviral vector that encodes an inhibitor of a HlV co-receptor, such as CCR5 or CXCR4, and a protein that inhibits HIV fusion to target cells and/or HIV replication. Pharmaceutical compositions comprising such constructs and methods of use thereof to prevent or treat HIV infection in a patient are also disclosed.


French Abstract

La présente invention concerne un vecteur d'expression pour la prévention ou l'inhibition de l'entrée, la fusion ou la réplication du VIH dans des cellules mammifères. En particulier, l'invention concerne un vecteur rétroviral recombinant qui encode un inhibiteur d'un corécepteur VIH, tel que CCR5 ou CXCR4, et une protéine qui inhibe la fusion du VIH pour cibler des cellules et/ou la réplication du VIH. L'invention porte en outre sur des compositions pharmaceutiques comprenant de telles constructions et sur des procédés d'utilisation afférents permettant de prévenir ou de traiter l'infection du VIH chez un patient.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS:
1. An expression vector comprising a first nucleic acid sequence encoding an
inhibitor of an
HIV co-receptor and a second nucleic acid sequence encoding a protein that
inhibits HIV fusion
to a target cell or HIV replication.
2. The expression vector of claim 1, wherein the expression vector is a viral
vector.
3. The expression vector of claim 2, wherein the viral vector is a lentiviral
vector or a
retroviral vector.
4. The expression vector of claim 3, wherein the lentiviral vector is self-
inactivating.
5. The expression vector of claim 1, wherein the expression vector confers
resistance to
infection by X4- and R5-tropic HIV strains when expressed in a host cell.
6. The expression vector of claim 1, wherein the expression vector confers
resistance to
infection by highly active antiretroviral therapy (HAART)-resistant HIV
strains when expressed
in a host cell.
7. The expression vector of claim 1, further comprising a third nucleic acid
sequence
encoding an inhibitor of HIV viral binding, HIV viral fusion to the target
cell membrane or HIV
viral replication.
8. The expression vector of claim 1, wherein the inhibitor of an HIV co-
receptor is a siRNA
or shRNA having a double-stranded region, wherein the double-stranded region
comprises a
sequence that is substantially identical and complementary to a sequence of
said HIV co-
receptor.
9. The expression vector of claim 8, wherein said HIV co-receptor is CCR5 or
CXCR4.
55

10. The expression vector of claim 9, wherein the shRNA has a sequence of SEQ
ID NO: 1.
11. The expression vector of claim 1, wherein the inhibitor is capable of
reducing expression
of the HIV co-receptor when said vector is expressed in a host cell.
11. The expression vector of claim 1, wherein the protein that inhibits HIV
fusion to a target
cell is a C46 protein, a T20 protein, enfuvirtide, CP32M, and sifuvirtide.
13. The expression vector of claim 1, wherein the protein that inhibits HIV
replication is
selected from the group consisting of human TRIM5.alpha., rhesus TRIM5.alpha.,
chimeric TRIM5.alpha., a
human TRIM5-cyclophilin fusion protein, cyclophilin, E3 ubiquitin, APOBEC3G,
and bone
marrow stromal cell antigen 2 (BST-2).
14. The expression vector of claim 13, wherein the chimeric TRIM5.alpha.
comprises an amino
terminal domain from a human TRIM5.alpha. protein and a carboxy terminal
PRYSPRY domain from
a rhesus TRIM5.alpha. protein.
15. The expression vector of claim 1, wherein said first and second nucleic
acid sequences
are operably linked to a promoter.
16. The expression vector of claim 15, wherein said promoter is a RNA
polymerase II or
RNA polymerase III promoter.
17. The expression vector of claim 1, wherein said first nucleic acid sequence
is operably
linked to a first promoter and said second nucleic acid sequence is operably
linked to a second
promoter.
18. The expression vector of claim 17, wherein said first and second promoters
are the same.
19. The expression vector of claim 17, wherein said first and second promoters
are different.
56

20. The expression vector of claim 19, wherein said first promoter is a RNA
polymerase III
promoter and wherein said second promoter is a RNA polymerase II promoter.
21. The expression vector of claim wherein said third nucleic acid sequence is
operably
linked to a third promoter.
22. The expression vector of claim 21, wherein said third promoter is the same
as or different
from the first and second promoters.
23. The expression vector of claim 16, wherein said RNA polymerase III
promoter is an H1
pol III promoter.
24. The expression vector of claim 16, wherein said RNA polymerase II promoter
is a
UbiquitinC pol II promoter.
25. A host cell comprising the expression vector of claim 1.
26. The host cell of claim 25, wherein the host cell is resistant to infection
by X4- or R5-
tropic HIV strains.
27. The host cell of claim 26, wherein the host cell is resistant to infection
by HAART-
resistant HIV strains.
28. The host cell of claim 25, wherein said host cell is hematopoietic
progenitor/stem cell, a
monocyte, a macrophage, a peripheral blood mononuclear cell, a CD4+ T
lymphocyte, a CD8+ T
lymphocyte, or a dendritic cell.
29. An expression vector comprising a first nucleic acid sequence encoding an
shRNA
having a sequence of SEQ ID NO: 1 and a second nucleic acid sequence encoding
for a C46
protein, wherein said first nucleic acid sequence is operably linked to an H1
pol III promoter and
said second nucleic acid sequence is operably linked to a UbiquitinC pol II
promoter.
57

30. An expression vector comprising a first nucleic acid sequence encoding a
first shRNA, a
second nucleic acid sequence encoding a second shRNA, and a third nucleic acid
sequence
encoding an inhibitor of HIV viral fusion to a target cell or HIV replication.
31. The expression vector of claim 30, wherein said first nucleic acid
sequence is operably
linked to a first promoter, said second nucleic acid sequence is operably
linked to a second
promoter, and said third nucleic acid sequence is operably linked to a third
promoter.
32. The expression vector of claim 30, wherein said first shRNA targets CCR5
and said
second shRNA targets CXCR4.
33. A method of treating or preventing HIV infection in a patient comprising
transducing
hematopoietic cells with the expression vector of claim 1 and transplanting
said transduced
hematopoietic cells in the patient, wherein said transduced hematopoietic
cells are resistant to
HIV infection.
34. The method of claim 33, wherein said hematopoietic cells are hematopoietic
progenitor/stem cells (HPSC), CD4+ T lymphocytes, CD8 + T lymphocytes,
monocyte/macrophages, or combinations thereof.
35. The method of claim 34, wherein said transplanted HPSC generate
granulocytes,
monocyte/macrophages, and lymphocytes that are resistant to HIV infection.
36. The method of claim 33, wherein said hematopoietic cells are autologous or
allogeneic.
37. The method of claim 33, wherein said first nucleic acid sequence encodes a
siRNA or
shRNA having a double-stranded region, said double-stranded region comprising
a sequence that
is substantially identical and complementary to a sequence of CCR5.
38. The method of claim 37, wherein the shRNA has a sequence of SEQ ID NO: 1.
58

39. The method of claim 37, wherein said transduced hematopoietic cells
express reduced
levels of CCR5 protein as compared to non-transduced hematopoietic cells.
40. The method of claim 33, wherein said second nucleic acid sequence encodes
a protein
selected from the group consisting of human TRIM5.alpha., rhesus TRIM5.alpha.,
chimeric TRIM5.alpha., a
human TRIM5-cyclophilin fusion protein and a C46 protein.
41. The method of claim 40, wherein the chimeric TRIM5.alpha. comprises an
amino terminal
domain from a human TRIM5.alpha. protein and a carboxy terminal PRYSPRY domain
from a rhesus
TRIM5.alpha. protein.
42. The method of claim 40, wherein said transduced hematopoietic cells
express increased
levels of said protein as compared to non-transduced hematopoietic cells.
43. The method of claim 35, wherein said granulocytes, monocyte/macrophages,
and
lymphocyte's are resistant to infection by R5 and X4 tropic strains of HIV.
44. The method of claim 43, wherein said granulocytes, monocyte/macrophages,
and
lymphocytes are resistant to infection by HAART-resistant HIV strains.
45. The method of claim 33, wherein the patient is naïve to HAART.
46. The method of claim 33, wherein the patient is receiving a HAART regimen.
47. The method of claim 33, wherein the patient is failing or has failed on a
HAART
regimen.
48. A pharmaceutical composition comprising an effective amount of the
expression vector
of claim 1 and a pharmaceutically acceptable carrier.
59

49. Use of a pharmaceutical composition of claim 48 in the manufacture of a
medicament for the treatment of a patient infected with HIV.
50. A method of treating or preventing HIV infection in a patient comprising
administering
the pharmaceutical composition of claim 48 to the patient.
51. The method of claim 50, wherein the patient is resistant to infection by
R5 and X4 tropic
strains of HIV following administration of the composition.
52. The method of claim 51, wherein the patient is resistant to infection by
HAART-resistant
HIV strains following administration of the composition.
53. The method of claim 50, wherein the patient is naïve to HAART.
54. The method of claim 50, wherein the patient is receiving a HAART regimen.
55. The method of claim 50, wherein the patient is failing or has failed on a
HAART
regimen.
56. The method of claim 50, wherein the patient is at risk for HIV infection.
57. Use of a viral expression vector of claim 1 in the manufacture of a
medicament for the treatment of a patient infected with HIV.
58. A method of producing a viral expression vector which, when present in a
cell, is
capable of inhibiting binding of HIV to the cell and preventing HIV fusion
into the
cell or HIV replication, the method comprising:
synthesizing a cDNA of a gene which expresses a protein capable of preventing
HIV fusion into a cell or HIV replication;
cloning the synthesized cDNA into a restriction site in a viral vector; and
inserting an expression unit capable of down regulating expression of an HIV
co-receptor
60

into a restriction site in the vector.
59. The method of claim 58, wherein the cDNA is a C46 cDNA or TRIM5.alpha.,
cDNA and the
expression unit is a shRNA targeting CCR5.
60. The method of claim 59, wherein the shRNA has a sequence of SEQ ID NO: 1.
61. The method of claim 59, wherein the viral vector is a FG11F lentiviral
vector.
62. The method of claim 61, wherein the C46 or TRIM5.alpha. cDNA is cloned
into restriction
sites BamHI and EcoRI of an FG11F vector.
63. The method of claim 62, wherein the shRNA is inserted between Xbal/Xhol
restriction
sites of the FG11F vector.
61

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
DUAL VECTOR F OR INH_IBI"1 ION OF HUMAN IMMUNODEFICIENCY VIRUS
CROSS-REFERENCE TO RELATED APPLICATIONS
10001] This application claims the benefit of priority of U.S. Provisional
Application No.
61/287,599, filed December 17, 2009, and U.S. Provisional Application No.
61/225,687, filed
July 15, 2009, both of which are herein incorporated by reference in their
entireties.
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY
[0002] The contents of the text file submitted electronically herewith are
incorporated herein by
reference in their entiret~v: A computer readable format copy of the Sequence
Listing (filename:
CALI 004 01WO SegList ST2S.txt, date recorded: May 26, 2010, file size 9
kilobytes).
FIELD OF TfIE INVENTION
1100}3] The present invention relates generally to the fields of molecular
biology and virology.
In particular, the invention relates to expression vectors useful in the
treatment and prevention of
1IIY infections.
BACKGRO N OF T11 INVENTION
100041 1-Human immunodeficiency virus (I-HIV) is the causative agent of
acquired
immunodeficiency syndrome (AIDS) in humans, which causes the immune system to
fail and
increases the probability of death due to opportunistic infections. HIV
infection is a major global
health problem as evidenced by its designation as a pandemic by the World
Health Organization.
Most people who are infected with HIV eventually develop AIDS, which claims
the lives of
more than one million people every year.
[0005] Antiretroviral therapies, such as ITAr'sRT (highly active
antiretroviral therapy), which
includes combinations of nucleoside analogue reverse transcriptase inhibitors,
protease
inhibitors, and aon-nucleoside reverse transcriptase inhibitors, have
dramatically decreased the
morbidity and mortality rate from HIV'/ <" IDS in regions of the world where
the therapy is
available. However, l _'V.<" 1 T does not cure or completely eliminate all the
symptoms of
HINT/AIDS. THA_ART is also associated with several side effects as well as the
emergence of I-TTV
I

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
strains that are resistant to the retroviral inhibitors. For these reasons as
well as the high cost of
HA-ART and need for strict adherence, such therapy can be relatively
ineffective for a large
number of patients. Thus, there is need in the art to develop improved
strategies for treating and
preventing HIV infection
SUMMARY OF TIIE II ELATION
[0006] The present invention provides a new therapeutic approach for treating
and/or preventing
1W infection in which two different steps in viral infection are targeted by
gene therapy. For
instance, the present invention provides a vector encoding an inhibitor of
viral entry into a host
cell and an inhibitor of viral fusion and/or an inhibitor of viral
replication. Accordingly, in one
embodiment, the present invention provides an expression vector comprising a
first nucleic acid
sequence encoding an inhibitor of an III' co-receptor and a second nucleic
acid sequence
encoding a protein that inhibits I-1IV fusion to the target cell or IIIV rep]
ication, wherein said
first nucleic acid sequence is operably linked to a first promoter and said
second nucleic acid
sequence is operably linked to a second promoter. The expression vector can be
a viral vector,
such as a retroviral or lentiviral vector. In sortie embodiments, the first
and second nucleic acid
sequences are transcribed from a single promoter. In a particular embodiment,
an internal
ribosorne entry site (lRES) is present upstream of the second nucleic acid
sequence.
10007 In certain embodiments, the expression vector further comprises a third
nucleic acid
sequence encoding an inhibitor of viral entry, viral fusion, or viral
replication. In some
embodiments, the third nucleic acid is operably linked to a third promoter.
other
embodiments, two of the three nucleic acid. sequences are transcribed from a
single promoter (i.e.
the first and second nucleic acid sequences or the second and. third nucleic
acid sequences). In
still other embodiments, all three nucleic acid. sequences are transcribed
from a single promoter.
One or more IRES can be present upstream of the second and./or third. nucleic
acid sequences.
[0008] In one embodiment of the invention, the first nucleic acid sequence of
the expression
vector encodes an inhibitory nucleic acid molecule, such as a siR A or shRINA,
that targets an
HIV co-receptor. In some embodiments, the siRNA or shRNA molecule comprises a
double-
stranded region having a sequence that is substantially identical and
complementary to CCR5. In

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
other embodiments, the siR-N--A or shRNA molecule comprises a double-stranded
region having a
sequence that is substantially identical and complementary to CXCR4.
[0009] In another embodiment of the invention. the second nucleic acid
sequence of the
expression vector encodes a protein that inhibits ITIV fusion to the target
cell. The fllY fusion
inhibitor protein can be a C46 protein or other like proteins that inhibit
fusion of IT1V to the cell
surface and are transgene expressed to be located on the cell surface (e.g.,
T20 and its related
proteins, enfuvirtide, CP32M, and situvirtide).
[0010] In yet another embodiment of the invention, the second nucleic acid
sequence of the
expression vector encodes a protein that inhibits 1IIV replication. For
instance, in some
embodiments, the second nucleic acid sequence encodes a TRIM5u, protein or a
derivative or
fusion thereof. In certain embodiments, the second nucleic acid sequence
encodes a chimeric
TRTM5u , in which the amino terminal domain is from a human TRIM5ox protein
and the carboxy
terminal PRY SPRY domain is from a rhesus TRTM5o: protein. In other
embodiments, the
second nucleic acid sequence encodes a TR_I M5-cy clophilin fusion protein.
100111 In one embodiment of the invention, the expression vector comprises a
first, second, and
third nucleic acid sequence, wherein the first nucleic acid sequence encodes
an inhibitor of an
lily co-receptor (e.g., sh R A to C,CR5 or C X(,R4), the second nucleic acid
sequence encodes a
fusion inhibitor (e.g., C46), and the third nucleic acid sequence encodes an
inhibitor of HIV
replication (e.g., TRI M5u protein or a derivative or fusion thereof).
[0012] In some embodiments of the invention, the inhibitor of an HIV co-
receptor and the
inhibitor of lily fusion to the target cell or inhibitor of I-1I replication
are expressed from
different prorn_oters on the expression vector. In one embodiment, the
inhibitor of an 111% co-
receptor (e.g. CCU or (,:X(='R4) is expressed from a RNA polyrnerase III
promoter, while the
inhibitor of II IV fusion and/or replication is expressed from a RNA
polymerase 11 promoter. The
two different inhibitors can he expressed in different ratios from the
expression construct.
3

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0013] The present invention also provides methods of making the expression
vectors described
herein as well as pharmaceutical compositions comprising the novel expression
vectors. For
instance, in one embodiment, the method of producing a viral expression vector
which, when
present in a cell, is capable of inhibiting binding of HIV to the cell and.
preventing HIV fusion
into the cell or I-l replication, comprises synthesizing a cDNA of a gene
which expresses a
protein capable of preventing HIV fusion into a cell or HIV replication;
cloning the synthesized
elit'd_A into a restriction site in a viral vector; and inserting an
expression unit capable of down
regulating expression of an HIV co-receptor into a restriction site in the
vector.
[0014] The present invention also provides a method of treating or preventing
HIV infection in a
patient. In one embodiment, the method comprises administering to the patient
a pharmaceutical
composition comprising an expression vector of the invention. Administration
of such
compositions can confer resistance to infection b RS and X4 tropic strains of
H [V. In one
embodiment, the patient is human. The patient may be UIV negative or UIV
positive. In some
embodiments, the patient may be naive to HAART therapy, receiving HAART
therapy, failing or
failed on UAART therapy. In other embodiments, the patient may have full-blown
AIDS (e.g.,
IDS/lyinphorna).
10015+1 In another embodiment', the method comprises transducing
hemnatopoietic cells (e.g.,
HIPSC,, CD4+ T lymphocytes, C D8+ I lymphocytes, or mnonocyte/macrophages)
with an
expression vector of the invention and transplanting said transduced cells in
the patient, wherein
said transduced cells are resistant to HIV infection. In one embodiment, the
hematopoietic cells
are hematopoietic progenitor/stem cells (HPSQ that generate granulocytes,
monocyte/macrophages, and lymphocytes that are resistant to HIV infection
following
transplantation into a patient. In some embodiments, the HI'SC are autologous
and CD34
positive. The transduced HPSC can generate granulocytes, monocyte/macrophages,
and
lymphocytes that are resistant to infection by R5 and X4 tropic strains of
HIV. In certain
embodiments, the transduced HPSC- can generate granulocytes,
monocyte/macrophages, and
lymphocytes that are resistant to infection by HIV strains that are resistant
to HA:'4I T.
4

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
]0016] Throughout this specification, unless the context requires otherwise,
the word
"comprise", or variations such as "comprises" or "comprising", will be
understood to imply the
inclusion of a stated element, integer or step, or group of elements, integers
or steps, but not the
exclusion of any other element, integer or step, or group of elements,
integers or steps.
[0017] Any discussion of docinvents, acts, materials, devices, articles or the
like which has been
included in the present specification is solely for the purpose of providing a
context for the
present invention. It is not to be taken as an admission that any or all of
these matters form part
of the prior art base or were common general knowledge in the field relevant
to the present
invention before the priority date of each claim of this specification.
100181 In order that the present invention may be more clearly understood,
preferred
embodiments will be described with reference to the following drawings and
examples.
BRIEF DESCRIPTION OF THE DRAWINGS
100191 Figure 1. Lentiviral Vector Constructs. Schematic showing the important
elements
comprising each of the indicated vectors. The dual vector sh5/C:46 is shown
highlighted by
dotted line surround.
10020] Figure 2. Backbone Constructs. Schematic showing the important elements
comprising
each backbone lentiviral vector. p, means plasinid. pFGI IF was obtained from
pFG12 by
inserting multiple cloning sites (MCS) at various locations including upstream
of the ubiquitin
promoter (Ubc).
]0021] Figure 3. Vectors Derived from F G12 Backbone. Schematic showing the
derivation of
pFGI2-Hi R5 W_EGFl' and pFG12.-HI-R5
]0022] Figure 4. Vectors Derived from FGIIF Backbone. Schematic showing the
derivation
of pFG I IF- UFC46 and pFG I I F-H I .R5-U- 0446.

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0023] Figure 5. Production of Lentivirus. Schematic (left panel) shows the
HIV-1 wild-type
genonae and the generic vectors used in the transient co-transfection system:
1. HIV vector
plasmid (test vector, c. g. constructs shown in Figures 1-4); 2-4 the various
helper plasmids.
Schematic in dashed box (right panel) shows the elements of the actual helper
plasmids used in
lentivira1 production.
[0024] Figure 6, Stability of Expression in CEM,NKR,CC 5 Cells. FRCS analysis
of
CEM.- KR.CCR5 cells transduced with the indicated constructs at 4 and 8 weeks
in culture.
Cells were analyzed for CCR5 expression (via CD195 antibody), C46 expression
(via. 2F5
antibody), and EGFP expression. GFP expression is seen for the constructs
containing EGFP
(GFP control and shy/ ECFP; panels 1,3); a reduction in CCR5 expression is
seen for the
constructs containing shy (shy, shy/l GFP, and sh5/C46; panels 2, 3,5), and
C46 expression is
seen for the constructs containing C46 (C46 and sh5/C46; panels 4,5).
Percentage positive cells
are shown in each flow cytometry quadrant (IQ 1-Q4). Similar results are seen
at 4 and 8 weeks.
100251 Figure 7. Stability of Expression in Molt4/CCR5 Cells. FAGS analysis of
Molt4/CCR5
cells transduced with the indicated constructs at 4 and $ weeks in culture.
Cells were analyzed
for CCR5 expression (via CD 195 antibody), C46 expression (via 2F5 antibody),
and EC&P
expression. (WI) expression is seen for the constructs containing EGFP ((FP
control and
sh5/E IFP; panels 1,3); a reduction in CCR5 expression is seen for the
constructs containing sh5
(shy, sh5/E IFP, and sh5/C46; panels 2,3,5), and C46 expression is seen for
the constructs
containing C46 (C46 and sh5/C46; panels 4,5). Percentage positive cells are
shown in each flow
cytoretry quadrant (Q1-Q4). Similar results are seen at 4 and 8 weeks.
[0026] Figure 8. Growth Characteristics of Transduced CEi4M,NKR,CCRS Cells,
Bar graph
showing the number of CEM.NKR.CCRS cells transduced with the indicated
lentiviral
constructs 4-7 days after seeding at the indicated concentrations; 4
independent seedings are
shown numbered on X-axis 1-4. Transduction with the various constructs
(sh5(2)o sh5/EGFP(3),
C46(4), sh5/C46(5)) had no effect on the growth rate of the cells compared to
untransduced
eells(1).
6

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
100271 Figure 9. Transduction Methods for Peripheral Blood Mononuclear Cells
(PBN C).
PBMC were transduced with a shy/EGFP lentiviral construct in one of the four
following ways:
lx transduction with virus containing medium (VCM), 2X transduction with VCM,
1X
transduction with'JCM preload (Preload x 1), 2X transduction with VCM preload
(Preload x 2),
and concentrated VCM. A. Flow cytometry analysis of PBMC transduced with
shy/EC FP
lentiviral construct with the indicated methods. B. Summary of the percentage
of EGFP positive
cells for each transduction method. The results show that transduction was
most efficient with
concentrated virus, followed by pre-load 2X, pre-load l x, then 2X and 1 X
suspension; two
replicates are shown for each.
[0028] Figure 10. PB MC Transduction. FACS analysis of PBMC transduced with
the
indicated constructs at 4 days post transduction. Cil/P expression is seen for
the constructs
containing E(-'3 FP (panels 1,2); CC R5 down-regulation is seen for the
constructs containing shy
(panels .2 and 4), and C46 expression (measured by 2F5 antibody) is seen for
the constructs
containing C46 (panels 3,4). MF _ values, from left to right, were 16.2, 8.4,
1 6.8, 9.4.
[0029] Figure 110 Comparison of gene expression in transduced PBMC (at day 4)
and
transduced CEM.NKR.CCR5 T cells (at week 8). Similar expression patterns are
observed
between the two cell types. GFP expression is seen in cells tra;nsduced with
the constructs
containing EGFP (panels 1,2). CCR5 expression is reduced in cells transduced
with the
constructs containing shy panels 2 and 4), and C46 expression (measured by 2F5
antibody) is
observed in cells transduced with the constructs containing C'46 (panels 3,4).
[0030] Figure 12. Growth Characteristics of Transduced Human Pi41C. Total
cells/well
(panel A) and percentage of viable cells (panel B) were similar for PBMC
transduced with each
of the indicated constructs and. PBMC that were not transduced. Two replicate
seeds of each
group are shown,
[0031] Figure 13. Stability of Transgene Expression in PBMC. FACE analysis of
PBMC
transduced. with the indicated. constructs at 4, 7 and 12 days post
transduction, GFP, CCR5, and

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
C46 expression (as measured by 2F5 antibody) were analyzed, GFP expression is
seen in panels
1,39 shy expression is seen in panels 293,5 and. C46 expression is seen in
panels 4,5.
[00321 Figure 14. CD34+ Isolation and Transduction. FACE analysis of human
mononuclear
cell populations before (pre-separation) and after (post-separation) isolation
of CD34-+- cells by
Magnetic Antibody Cell Separation (upper panel). FACE analysis of human CD34
hematopoietic stein cells transduced with the indicated constructs (bottom
panel). GFP
expression is seen in panels 1,2, C46 expression is not seen in panels 4,5,
[00331 Figure 15. HIV Challenge with Dual Tropic SF2 Strain in olt4/CCR5
Cells.
Mo1t4/CCR5 cells were either non-transduced or transduced with sh5/C46
lentiviral vector and
subsequently challenged with 1-. l -SF2 dual tropic (CCR5 and C='X(;R4) virus
at varying
multiplicity of infection (MOI) 0.2, 0.02, 0.002. P24 protein levels were
assessed 13 days after
viral challenge as a measure of HIV infection.
100341 Figure 16. 11V Challenge with Dual Tropic SF2 Strain in Molt4/CCRS
Cells.
NM1_olt4/CCRS cells were either non-transduced or transduced with sh5/C46 or
C46 lentiviral
constructs and subsequently challenged with TI1V-SF2 dual trope.. (CCR5 and
CXCR4) virus at
two different multiplicity of infection (MOO) -- 0.2 and 0.02. 124 protein
levels were assessed 11
days after viral challenge as a measure of HIV infection (upper panel). FACE
analysis of non-
transduced Molt4/CClR5 cells or Molt4/CCR5 cells transduced with C46 or
sh5/C46 lentiviral
constructs on the day of viral challenge (lower panel). CCR5 and C46 (as
measured by 2F5
antibody) expression was assessed.
10035 Figure 17. HIV Challenge with Dual Tropic SF2 Strain in Molt4/CCI Cells.
Molt4/CCR5 cells were either non-transduced or transduced with C46 (Gene 2.)
or sh5/C46
(G2R5) lentiviral constructs and subsequently challenged with HIV-SF2 dual
tropic (CCR5 and
CXCR4), Bal (CCR5 tropic) or NL4-3 (CXCR4 tropic) virus at an MOI of 0.2.. P24
protein
levels were assessed 11 days after viral challenge as a measure of HIV
infection. The numbering
on the histograms (1-6) refers to the HI\' strains that were used (see key on
right-hand side).
8

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0036] Figure 18. HIV Challenge with CCRS Tropic Bal Strain in Molt4!CCR5
Cells.
Molt4/CCR5 cells were either non-transduced ( Iolt4) or transduced with one of
the following
four lentiviral constructs: shy (R5); C46 (G2); sh5/C46 (R5-G2.); or shy./EGFP
(K5-GFP). The
nix" group is a mixture of untransduced, shy. C46, sh5/C46 all mixed equally
(i.e. 25''0 of each
type). The cells were subsequently challenged with HIV-Bal CCR5 tropic virus
at a multiplicity
of infection (MOT) of 022. P24 protein levels were assessed 7 and 10 days
(first and second
histogram respectively for each treatment) after viral challenge as a measure
of HIV infection.
[0037] Figure 19. IIIV Challenge in Peripheral Blood Mononuclear Cells. A.
Diagram of
Dual sh I 005/C46 Construct. B. PBMC were trransduced with one of the
following four lentiviral
constructs: sh5/C46 (LVsh5C46); C46 (LVC46); shy/GFP (LVsh5-C FP); or G FP
control (LV-
GFP). FACS analysis of transduced PIIMC four days post transduction. C.
Sixteen days post
transduction, cells shown in panel 13 were challenged with either a CCRS (R5)-
tropic or CXCR4
(:X4)-tropic HIV strain and p24 protein levels were assessed in culture
supernatants four days
following viral challenge.
[003$] Figure 20. Efficient CCR5 reduction in the NOD SCID-hu BLT mouse. A.
Flow
Cytuuaetryy. The percent CCE5 expression in EGFP+- (upper panel) and rnCherry4
(lower panel)
CD4 T-cells was examined by FACS analysis in lymphoid organs of reconstituted
mice.
Representative data from a mouse at 20 weeks post reconstitution is shown.
Thy/Liv:
Transplanted human thyrnus like organoid. LPL: Lamina propria lymphocytes. B.
CCRS tropic
H IV4I inhibition ex vivo. Splenocytes isolated from a transplanted mouse were
activated with
PHA for 2 days and IL-2 for 5 days and CD8+ cells were depleted. Cells were
sorted for EGFP+
and mCherry+ at over 99% purities. Sorted EGFP (black diamond) and mCherry+
(open
square) cells (4 x 104) were infected with RS HIV_ INTNSXsr,9 or X4 HIV INi,4-
; at MGl of 2.5 in
parallel and in triplicate. Cells were washed 3 times after the infection. The
amount of remaining
input HIV-1 particles in culture supernatant was monitored I hour after
infection by HIV p24
ELISA assay. The amount of HIV production in culture supernatant was monitored
at day 4, 7
and 12 after infection during the culture. C. Selective advantage of CCR5
dowuregulated
CD4+ T-cells in vivo. Reconstituted mice were infected with R5 tropic IIIV I -
FNSXSL9
(dose=200 me of p24) at 9 weeks post HPSC transplant. Kinetics of % EGFP+ CD4+
T-cell
9

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
population (Gray bar) in peripheral blood was monitored for 8 weeks after R5
tropic HIV
injection. %% mCheriv+ CD4+ T-cell population (White bar) was monitored within
the same
animal. The % EGFP+ and % mCherry+ were maintained in HIV uninfected. mice at
17 week
post HPSC transplant (data not shown). Representative data is shown. D.
Selective maintenance
of CD4/CD8 ratio in vivo. Kinetics of CD4,/CD8 ratio in EGFP+ CD45+ T-cell
population
(Gray bar) in peripheral blood was monitored during 8 weeks after R5 tropic
HIV injection.
CH4/CD8 ratio in % mCherry+ CD4+ T-cell population (White bar) was monitored
within the
same animal, A representative animal is shown. The CD4/CD8 ratio in Ef 3FP+
and mCherry+
CH45+ cells were maintained above I in lIl uninfected mice at 17 week post
HPSC transplant
(data not shown).
[0039] Figure 21, Predicted impact of introducing 05/C46-transduced 0034+
and/or
C04+ cells into an ]-1IV+ individual naive to HART. Predicted viral load (A)
and CD4
count (B) in patients treated with one dose of transduced cells (stars) versus
untreated patients
(triangles).
[0040] Figure 22. Predicted impact of introducing sh5/C46-transduced C D34+
and/or
CD4+ cells into an 14[V+ individual on a well-con trolled HAART regimen. The y-
axis
depicts predicted viral load. The x-axis details when antiretroviral therapy
(Awl') is being taken
or when an analytic treatment interruption (All) is instituted. Predicted
viral loads for patients
treated with one dose of transduced cells (stars) versus untreated patients
(triangles) is shown.
10041] Figure 23. Predicted impact of introducing sh5/C46-transducÃd C D34+
and/or
C04+ cells into an HIV+ individual failing HA-/RRT. Predicted viral load (A)
and CD4 count
(B) in patients treated. with one dose of transduced cells (stars) versus
untreated patients
(triangles
[0042] Figure 24. Dual Lentiviral Construct with shRNA targeting CCR5 and a
TRIMScx
protein. A. Schematic showing the elements of pFG11F-1-I1-R54 -TRI 15ct. B
Schematic
showing the derivation of the triple vector pFG1 I F-1-11-R5-U-C46-IB-
TRIM_Sa..
1 l1

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
DETAILED DESCRIPTION OF THE INVENTION
[00431 The present invention is based., in part', on the recognition that a
therapeutic approach that
targets non-HIV genes and/or proteins (that is host cell genes and/or
proteins) decreases the
probability that new HIV strains resistant to the inhibitors will emerge. In
particular, the present
invention provides vectors and methods of using such vectors to prevent or
treat HIV infection
by targeting or employing cellular proteins that affect different stages of
the HIV life cycle, For
instance, in one embodiment, the vector is capable of expressing an inhibitor
of viral entry
(binding) and an inhibitor of viral fusion to the cell membrane. In another
embodiment, the
vector is capable of expressing an inhibitor of viral entry and an inhibitor
of viral replication.
Accordingly, the present invention provides an expression vector comprising a
first nucleic acid
sequence encoding an inhibitor of an I-TTV co-receptor and a second nucleic
acid sequence
encoding a protein that inhibits HIV viral fusion to a target cell or IV
replication.
100441 In one particular embodiment, all three elements (e.g., an inhibitor of
an HIV co-
receptor, a protein that inhibits I.11V viral fusion to a target cell and a
protein that inhibits HIV
replication) are combined in one vector. For instance, in one ermrbodirnent,
the expression vector
comprises a first, second, and third nucleic acid sequence, wherein the first
nucleic acid sequence
encodes an inhibitor of an I-1IV co-receptor (e.g., shRNA to CCR5 or CXCR4),
the second
nucleic acid sequence encodes a fusion inhibitor (e.g., C46), and the third
nucleic acid sequence
encodes an inhibitor of HIV replication (e.g., TRlM5c. protein or a derivative
or fusion thereof,).
In another embodiment, the expression vector comprises a first, second, and
third nucleic acid
sequence, wherein the first nucleic acid sequence encodes a first inhibitor of
an I-11% co-receptor
(e.~. shRNA to CCU), the second nucleic acid sequence encodes a second
inhibitor of an Her
co-receptor (e.g., shRNA to C~X(:R4), and the third nucleic acid sequence
encodes an inhibitor of
IIIV viral f ision to a target cell (e.g., 045).
F0045I As used herein, "expression vector" or "vector" refers to a composition
of matter which
can be used to deliver nucleic acids of interest to the interior of a cell
such that they will be
expressed by the cell. Numerous vectors are known in the art including, but
not limited to, linear
polynucleotides, polynucleotides associated with ionic or amphiphilic
compounds, plasrnids, and
viral vectors. Examples of viral vectors include, but are not limited to,
adenoviral vectors,
11

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
adeno-associated virus vectors, retroviral vectors (including lentiviral
vectors), and the like. In
one embodiment, the expression vector is a viral vector. Preferably, the viral
vector is a
retroviral or lentiviral vector.
[0046] "Retroviruses" are viruses having an RNA genome that is reverse
transcribed by
retroviral reverse transcriptase to a cDNA copy that is integrated into the
host cell genome.
Retroviral vectors and methods of making retroviral vectors are known in the
art. Briefly, to
construct a retroviral vector, a nucleic acid encoding a gene of interest is
inserted into the viral
genoine in the place of certain viral sequences to produce a virus that is
rep] I cation-defective, In
order to produce virions, a packaging cell line containing the gag, pol, and
env genes but without
the LTR and packaging components is constructed (Mann et a1., Cell, Vol.
33.153-159, 1983).
When a recombinant plasmid containing a cDNA, together with the retroviral LTR
and
packaging sequences, is introduced into this cell line, the packaging sequence
allows the RNA
transcript of the recombinant plasmid to be packaged into viral particles,
which are then secreted
into the culture media. The media containing the recombinant retroviruses is
then collected,
optionally concentrated, and used for gene transfer (see Example 1).
100471 "Lentivinis" refers to a genus of retroviruses that is capable of
infecting dividing and non-
dividing cells. Several examples of lentivimses include HIV (humnan
immunodeficiency virus:
including 1-11 V type 1, and HIV type 2,), the etiologic agent of the human
acquired
immunodeficiency syndrome (AIDS); visna-mnaedi, which causes encephalitis
(visna) or
pneumonia (Fnaedi) in sheep, the caprine arthritis-encephalitis virus, which
causes immune
deficiency, arthritis, and encephalopathy in goats; equine infectious anemia
virus, which causes
autoimnrune hemolytic anemia, and encephalopathy in horses; feline
immunodeficiency virus
(P'It), which causes immune deficiency in cats; bovine immune deficiency virus
(BIV), which
causes lymphadenopathy, lymphocytosis, and possibly central nervous system
infection in cattle;
and. simian immunodeficiency virus (SIV), which causes immune deficiency and
encephalopathy
in sub-human primates.
[0048] A "hybrid virus" as used herein refers to a virus having components
from one or more
other viral vectors, including elements from non-retroviral vectors, for
example, adenoviral-
12

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
retroviral hybrids. As used herein hybrid vectors having a retroviral
component are to be
considered within the scope of the retroviruses.
[00491 A "pseudotyped" retrovirus is a retroviral particle having an envelope
protein that
is from a virus other than the virus from which the RN A genome is derived.
The envelope
protein may be from a different retrovirus or from a non-retroviral virus. A
preferred envelope
protein is the vesicular stomatitis virus G (VSV U) protein, However, to
eliminate the possibility
of human infection, viruses can alternatively be pseudotyped with ecotropic
envelope protein
that limit infection to a specific species, such as mice or birds. For
example, in one embodiment,
a mutant ecotropic envelope protein is used, such as the ecotropic envelope
protein 4.17 (Powell
e/ al. Nature Biotechnology 18(12):1279-1282 (2000)).
10050] The term "provirus" is used to refer to a duplex DNA sequence present
in a eukaryotic
chromosome that corresponds to the genome of an RNA retrovirus. The provirus
may be
transmitted from one cell generation to the next without causing lysis or
destruction of the host
cell,
100511 A lentiviral genorne is generally organized into a 5 long terminal
repeat (LTR), the gag
gene, the pol gene, the env gene, the accessory genes (nef, vif, vpr, vpu) and
a 3' LTR. The viral
LTR is divided into three regions called U3, R and US. The U3 region contains
the enhancer and
promoter elements. The U5 region contains the polyadenylation signals. The R
(, repeat) region
separates the U3 and US regions and transcribed sequences of the R region
appear at both the 5'
and 3' ends of the viral RNA. See, for example, "RNA Viruses: A Practical
Approach" (Alan J.
Cann, Ed. , Oxford University Press, (2000))9 0 Narayan and Clements (1989) J.
Gen. Virology,
Vol. 70:1617-1639; Fields et al. (1990) Fundamental Virology Raven Press.;
Miyoshi H. Blamer
U. Takahashi M. Gage F H, Verma I M. (1998) J Virol., Vol. 72(10):8150 and
U.B. Pat. No.
6,013,516.
[00521 Lentiviral vectors are known in the art, including several that have
been used to infect
hematopoietic progenitor/stem cells (HPSC). Such vectors can be found, for
example, in the
following publications, which are incorporated herein by reference: Evans et
al., flung Gene
13

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
Ther., Vol. 10:1479-1489, 1999; Case et a/., Proc Natl Acad Sci USA, Vol.
96:2988-2993, 1999;
Uchida et al., Proc Natl Acad Sci USA, Vol. 95:11939-11944, 1998; Miyoshi et
al., Science,
Vol. 283:682-686, 1999; and Sutton et al., J. Virol., Vol. 71".:5781-5788,
1998. In one
embodiment, the expression vector is a modified lentivims, and thus is able to
infect both
dividing and non-dividing cells. Such lentiviral vectors comprise a modified
lentiviral genome
that comprises a first nucleic acid sequence encoding an inhibitor of an HIS'
co-receptor and a
second nucleic acid sequence encoding a protein that inhibits I-l fusion to a
target cell or HIV
replication. Further, the modified lentiviral genome preferably lacks genes
for lentiviral proteins
required for viral replication, thus preventing undesired replication, such as
replication in the
target cells. The required proteins for replication of the modified genome are
preferably provided
in trans in the packaging cell line during production of the recombinant
retrovirus (or specifically
lentivirus). In one embodiment, the packaging cell line is a 293T cell line.
The lentiviral vector
preferably comprises sequences from the 5' and 3' long terminal repeats (LTRs)
of a lentivirus. In
one embodiment, the viral construct comprises the R and U5 sequences from the
5' LTR of a
lentivirus and an inactivated or self-inactivating 3` LTR froorn a lentivirus.
The LTR sequences
may be LTR sequences from any lentivirus including from any species or strain.
For example,
the LTR may be LTR sequences from I IIV, simian immunodeficiency virus (SIV),
feline
imnrunodeticiency virus (FIV) or bovine immunodeficiency virus (IBIV).
Preferably the LTR
sequences are HIV LTR sequences.
100531 In certain embodiments, the lentiviral vector comprises an inactivated
or self-inactivating
3' L'1"R-that is the lentiviral vector is self-inactivaten . A "self
inactivating 3' LTR` is a 3' LTR
that contains a mutation, substitution or deletion that prevents the LTR
sequences from driving
expression of a downstream gene, A copy of the U3 region from the 3' L'I'R
acts as a template
for the generation of both LTRs in the integrated provirus. 'Thus, when the 3'
LTR with an
inactivating deletion or mutation integrates as the 5' LTR of the provirus, no
transcription from
the 5' LTR is possible. This eliminates competition between the viral
enhancer/promoter and any
internal enhancer/promoter. Self inactivating 3' L'I'Rs are described., for
example, in Zufferey et
al., J. Virol., Vol. 72:9873-9880,1998; Miyoshi et al., J. Virol., Vol.
7z2:8150-8157, 1998; and
Iwakuma et al., Virology, Vol. 2.61:120-132, 1999. The 3' LTR may be made self-
inactivating by
any method known in the art. In one embodiment the U3 element of the 3' LTR
contains a
14

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
deletion of its enhancer sequence, preferably the T ]IA box, Spl and NF-kappa
B sites. As a
result of the self inactivating 3' LTR, the provims that is integrated. into
the host cell genome will
comprise an inactivated 5` LIT. The viral expression vectors of the invention
preferably do not
inhibit vector production in producer cells. In certain embodiments, the viral
expression vector
substantially lacks toxicity to transduced and gene-containing cells.
[00,54] The expression vector of the invention comprises a first nucleic acid
sequence encoding
an inhibitor of an HIS' co-receptor. In one embodiment, the HIV co-receptor is
CC chemokine
receptor 5 (CCR5). CC-R5 is the primary 111V- -I co-receptor for macrophage
tropic strains and is
essential for HIV infection. Population genetic studies have demonstrated that
individuals
homozygous for a defective CCR5 gene (e.g. C,CR5A32 ) are protected from HIS'
infection.
Interestingly, heterozygous individuals who exhibit a 50% reduction of CCR5 on
cells have a
substantially reduced disease progression rate. Individuals who are homozygous
for the
CCR5A32 allele appear to be normal except for an increased susceptibility to
West Nile virus
encephalitis. A small molecule CCR5 inhibitor, Maraviroc has been approved by
the FDA for
use in humans. This inhibitor is effective in preventing HIV-I infection and
although some
adverse effects were noted, there did not appear to be any such effects
resulting from blocking
CCR5 itself. As expected, HIV-1 resistance does occur, however, interestingly,
the major form
of resistance appears to be HIV- I variants that adapt to use the drug-
occupied form of CCR5
rather than CXC chemokine receptor 4 (CXCR4) or other co-receptors. Thus,
knockdown of
CCR5 (such as with siRN , shRNA, or antisense) may he more effective than
blocking access.
In another embodiment of the invention, the HIV co-receptor targeted is CXCR4,
which is the
major co-receptor for T-cell tropic strains.
[005] In certain embodiments, the inhibitor of an HIV co-receptor is an
inhibitory nucleic acid.
As used herein, an inhibitory nucleic acid includes, but is not limited to, a
small interfering RNA
(siRr A), a short hairpin RNA (shRNA), an aptamner, a ribozyme, and an
antisense
oligonucleotide. Thus, in one embodiment, the first nucleic acid sequence
encodes an inhibitory
nucleic acid that targets an HIV co-receptor. "Target" refers to the ability
of the inhibitor to bind
to and/or interfere with an endogenous transcript encoding the HIV co-
receptor. For instance, the
inhibitory nucleic acid can have a sequence that is substantially
complementary to a nucleic acid

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
encoding the HIV co-receptor such that the inhibitory nucleic acid binds to
the HIV co-receptor-
encoding nucleic acid thereby blocking the expression or initiating the
degradation of the co-
receptor nucleic acid. Accordingly, in some embodiments, the inhibitor of an
HIV co-receptor is
capable of reducing expression of the HIV co-receptor when the expression
vector encoding said
inhibitor is expressed in a host cell.
[0056] A "srnall interfering RNA" or "siRNA" is a double-stranded RNA molecule
that is
capable of inhibiting the expression of a gene with which it shares homology.
The region of the
gene or other nucleotide sequence over which there is homology is known as the
"target region."
In one embodiment the siR- :A may be a "hairpin" or stem-loop RNA molecule
(shRNA ),
comprising a sense region, a loop region and an antisense region complementary
to the sense
region. In other embodiments the siRNA comprises two distinct RNA_ molecules
that are non-
covalently associated to form a duplex.
[0057] In some embodiments, an expression vector of the invention comprises a
first nucleic acid
sequence encoding an antisense oligonucleotide having a sequence that is
substantially
complementary to at least a portion of a nucleic acid sequence encoding an
111V co-receptor,
such as (-'(-'R5 and/or C,:XC R4. As used herein, "substantially
complementary" refers to a
sequence that is at least about 8th%, 85%X,,, 90%, 95%, 96%, 9-1,1/,/0,,
981io. or 99%
complementar to a target polynucleotide sequence. In one embodiment, the
antisense
oligonucleotide has a sequence that is 100% complementary to at least a
portion of a nucleic acid
sequence encoding CCR5 or CXCR4. The antisense oligonucleotide can be from
about 15 to
about 30 nucleotides in length, and in some embodiments about 19 to about 25
nucleotides in
length,
[0058] In other embodiments, an expression vector of the invention comprises a
first nucleic acid
sequence encoding a siR- A or shRNA. The sill. or shR`~1 A preferably has a
double-stranded
region comprising a sequence that is substantially identical and complementary
to a portion of a
nucleic acid sequence encoding an HIV co-receptor, that is at least about 75%,
80%, 85%, 90%,
95%, 96%. 97%, 98%, or 99% identical and complementary to a portion of a
nucleic acid
encoding CC R-5 or C XC R4. In one embodiment, the siRNA or shRNA has a double-
stranded
16

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
region comprising a sequence that is 100% identical and complementary to a HIV
co-receptor
sequence (e. g. CCR5 and/or CXCR4). The double-stranded region of the s1R_N1_A
or shRNA can
be from about 5 to about 60 nucleotides in length, preferably about 10 to
about 30 nucleotides in
length, more preferably about 15 to about 25 nucleotides in length, such as
about 20 nucleotides
in length. In certain embodiments, the first nucleic acid sequence of the
expression vector
encodes a shR- :A having a stern-loop structure, wherein the stern or double-
stranded region is
substantially identical and complementary to a sequence of CCR_5 or CXCR4. The
loop region
of the shRNA can comprise from about 2 to about 15 nucleotides. In one
particular errrbodirnent,
the first nucleic acid sequence encodes a shRNA comprising a sequence of 5 -G
GCAAGCUC
- . '~r, G, CG õV -r'r r Irrr, 11 1 E~G''_]UU[?C. rC a);r C Al_'.~ G.[j}AAA`
1_, G A -,V'(~v _,_,l a 3 (SE Q ID NO: 1)[00591 The expression vector of the
invention preferably comprises a second nucleic acid
sequence encoding a protein that inhibits HIV fusion to a target cell or HIV
replication. In some
embodiments, the protein that inhibits HIV fusion to a target cell is a C46
protein. C46 is a
membrane anchored fusion inhibitor derived from the C-terminal heptad repeat
of HIV gp441
fused with a human inumunoglobulin hinge region and a CD34 transmenrbrane
domain. C46 is a
potent I-TIV fusion inhibitor, in a sense analogous to the FDA approved
soluble drug enfuvirtide
(T20) and acts at a point in the HJV life cycle distinct from CCRS co-receptor
attachment. In one
embodiment, the safety of C46 was tested in a phase I clinical trial in which
patients received an
infusion of autologous T-cells transduced with C46 retroviral vector. In
another aspect, the
patients had no gene therapy related adverse effects and did not develop
apparent anti-C46
immune reactions. In one embodiment, the second nucleic acid sequence encodes
a x:46 protein
comprising a sequence of,
P GrAGhTGRAP DGPPLLLLLL LG\iS GL i.S :^7[`E:^7D?E Td?} YTSLT SL_'_ ESQNQQEH
}:_ ]. .. KWA SLIVU 1:E K "iT[ _:E?`P APPV Ct. ].~'1 ." A].C1hV
~.R.SWS;P'T}['. T,'-'] 'P (SEQ ID NO: 2}.
In another embodiment, the second nucleic acid comprises a sequence of.
5' ~'I'GGC;AGCAG UGC SAC. GG AE GC;GC}?:a'a'; G.- GGACCAC GGCTGC'IGC'a
GC`.CUC` GC` G
CTGG"CG1 A ''G G C' LLrALL. G!-~TtjGGAGm GL--Pa.. C,. Gv--P. r,' ~~h' ~~r'
'v-,.. ~. .. "Lr'., l'.j- :~ c'~T' ra-.. c'~h'
TA r ,L, iL c3 r "T GAT 'v1-1 jc3'v v
A '.j - '. A-GA E r11 -L r
h ,A T _ ,vULCT LCrU C AA Cr'1LCrtj CC ACrT'.j "_' =t.~A P,1--_,L-_TI.C,
C.P,'v-'-.L%3v
-r"
:r : vr 1, l-r,~ !_.cr(_ !- ( ,Etj'.T'h `- Etj '." .nTyr; .nTyr.; .AC .' -r l_-
-r GA C l: l '.cr-
E1!- . - v C~
1 i

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
C"_'C;C;`]'GACCA .;CG:;AGCC Ca G:. 'G.;C'C',CG CDC *-'G( CA ~t t,'aAC'a`['
.:C"]'G1~']': AAC
C.;C C.;C:aGC;']' GG .GC. CCAC . GGC'GAGCGG . `C GC AGC, .AGC `.'G i-3' (SECS
i_D N0: 3).
Other suitable proteins that inhibit FIIV fusion to a target cell and can be
encoded by the second
nucleic acid sequence in the expression vectors of the invention include T20
and its related
proteins, enfuvirtide, CP32M, and sifu.virtide.
100601 an certain embodiments, the second nucleic acid sequence encodes a
protein that inhibits
1-1 IV replication. In some embodiments, the protein that inhibits 11I
replication is a tripartite
motif -containing 5 alpha (TI 'I5u) protein or derivatives or fusions thereof.
For instance, the
second nucleic acid sequence can encode a human TRIM5u, rhesus TRIM5a, a
chimeric
TR1M5a, or a human TRIMS-cyclophilin fusion protein. In one embodiment, the
second nucleic
acid sequence encodes a human T 15u, protein comprising a sequence of.
MASG L , i vK _.. VTC`P TCLE LLT Q?LSLDC GHSFCQACLT' ANHKKSMLDK GES SC 'vCPT
S` QPE !I`Pi1 RH.%ANI :%L,<T ~~EV~<iLSPEGQ KVDHCARHGE KLLLFCQEDG KVICWLCERS
QEHRGHHTFL __E TAREY `' KL AAT iLR QKQ'QWAEEELE ADIREEE-HS KTQ YD-_KTN
VLAJ T ] - W -'S, r -iv]-,-1.K t'- 1_~ .,-t1 c c- v ~7_ Q,-,_.,_[1n ' -,_[. -s
); -
I.~_[1riJ _._a ~~t': ~;__:__1.~]_
GSVmii õI QC=i; ]DG R SV TT,KK,~ TFPK :~QSRV RAP[) LKGI ~:V R,_1 T,']'DVR-IYW]-
l
'~ SF'1 R`~ S SP Jj) rQ1 i'Cv7 ''Y- rr - S` SI -S ~r K}Y
1'VA iJ.]_ 1. r.~~ .] .K ti` "l ~_] _[`l G . ."~~ :J 't 1 . 'h] (i ~: ....
ti':[ V 1111VS _'K K[., . `~dT_].GVCA G FQ P]DAL'4C CE KS I NXQPKYC:YW VI [ I
E r S~ ?QDS -; [.
PSVPFIIV p _ S VI ]-r pr RVGV CV E N_] Gu'a, [Y-K} S.SL S QPV``PY_~NPR
ax,G PMTLCS 'SS (SEQ [D NO: 4).
In another embodiment, the second nucleic acid sequence comprises a sequence
of.
S'~AT C~ õ~ C CI'GG'' TAATGTAAAG GAGGAG'T'~GA C ti'CC -'C'~CT'~'G"
G~. ATC rti' 'G ULT ~A
-` ' - AAC " C(_1`J_'G-ACTGC ter, CAC r.. r, _ P T, AT-1G, r,
! C l_! r.~ ' h! ~ r.~ ' l '].'G cr1. - C '`- ' '`G'' - ~r1 ~n GC C ry' '].'
h C ;A A Ij" 1 . nA,~~. n~.~,~~..r ~. ~~['! h~.,1 'h~.,, l_'].. ~~['
cr~['vl,.~_,v r 11
C ~, '` r -r.- -,~, '` _ Avi~,~, '` ,~~~, . r TA RT .~~ . r A C APB A'].I T,~,
G G T C; ~~ ~, 1
,~~~.]. ~, "I. ~ ~ ; 'r..G _ . ~. : : ~~ P. ~ ~ ~~~ ~~ . '. C" ~,. ~., ." CT
,:
AG GGA GGT ,A AGTTGAGC' AG ;(; ;cAc; A aAC,'I":[ l"~zk':[C A`_ IG'L CA ..(,A1
GAGA
AAAC'I":'C'I'AC; T'C'T':'C'I' 'ICA GGAGGACGGG AAGG`.CCA'i'T" GC:'GGC'rI'i'G G
.GC:
C;A ;GA. ;C;A ,(' .G`['G.G`i'CAC A A ,AGA:;G A:;G T`_. CC, ,GG 7A
GTAC,t`A~~.G`]':G
AAC;.[' _ ,A(_; ; C:ACGC, G !"j CAA'-' AGC ,GC_ AGGAAG.GA !~j ;r~ G I .[ ' GA-
. _ !~ !~
G _ õt~ GT'~ CA `T-~A~A~:C..At:'TGA .P ~T .P ~ G ~~AC]~A Ar"~T ~GAT ~
A~~G:I"1'Gt.'I'G G AAGACTCAAA TA .T~~~'l~T GP ~ ..A:.
GT'CTTGGC'AG A'T'I IGAGCA ACTGAGA_GAC ATCC:TGGAC' GGGAGGAGAG CAAT'GAGCTG
T r.'' GGA G".~-
vAA~A, vT ;G ~GAr~G~~r~G ~~ ' ~" .A.~At.A ,'1' ~:'1'~~;AAAA ~ tip: T`_ v~. A.'
i_ I'GAA A C
~:'I'GA.;
is

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
A'._.'GG'GC \G(_i?.GAC~?.(_~']- C',CTG~?_~.~i1-1.- , C,T'.!'~iTC._'C G
A'._'CTGGAGCA I _,GGCTGCAG
GGGTC-'AGTGA PGGAG_''! GC" TCAGGGTG'TG GATGGCGTCA TAAAAAGGAC GGAGAACGTG
AC::TT;AA GA A GTC..AG' A AAC T'"_ 11'C..CAA AA AATCAAAGGA GA G TG4'i'TT .G A
GC '1'CC 'i'GT 'i'
C' 'GAAAGGAA PGC 'AGAAG' GT 'TAGAGAG CTGACAGATG 'I'C:CGACG ti=A CTGGGT PGAT.
GT-GACAGTGG C'T;: C'AAA .AA T1AT'T T .ATGT GCTGT C'ATTT _ -AA GATAA GAGA .AA
,TG
ACj T "-'T- C~h - ~'A G (--'G AGGG U . CU. AGU. ACCT GACATTTGTG
C, ,.;~A h A .,. A .,~.~' Ah~`T'AT'
G'~ ~_TAhC_T GG ;A T-C,CT GGC, TC,T -AAA -TA T :-AATh z G~.h7A .h~' z'Th~ 7 C
~.ATTTC.~._TT UTT~..,(~.~ . U~.,(~~: .~.,
TGGGAGGTAG ACGTGTCCAA GAAAACTGCT TGGATCC"TGG GGGTATGTGC TGGC.TTr CAA
C ~... .[. .. ._!~<<j _ -11.[.i-A~1 1; G GG 1A.ttG1 J, i!~j,~ <jttl_
-r - G Gr"' " -~ 'A'].'Ahv ~~[ ~'!\IJA IJA l~G["].r.~[? ti~['h']~
GIA -1
T'_ ,CTAGA T ATGA GC:'i'TG~ l_(IT'1'CAA'TA TCACAAAC: ,A a'GGICFTTCTC
A'--'L'( 1A`['AAG'1 _ ,]- IC'1 CACTC,
'T'_'CT'_ a TTCT .:AGCCTG`['Aa' P CA`i C.
Ar"j k`.['G'a GG ~G a'C'C111 C. \ T'~; " 7.'CTG'T'~__ C'TC': CC AG. _'C'I_. GA-
3' (SEQ ID NO: 5).
100611 A "chimeric TRI1M5a" refers to a TRIM5a protein comprising domains or
fragments
from TRIM5a, proteins from two or more species. For example, a chimeric
TRIM5a, can
comprise at least one domain from a human TRII!!ISa and. at least one domain
from a rhesus
TRIN15a. In some embodiments, a chimeric TRTM5a protein comprises an amino
terminal
domain from a human TRIM5cx protein and a carboxy terminal PRYSPRY domain from
a rhesus
TRIM5cm protein.
100621 In another embodiment, the second nucleic acid sequence encodes a
fusion protein
comprising TRIM Sa and cyclophilin. For instance, in one embodiment, the TI
IM5-cyclophilin
fusion protein comprises amino acids I to about 309 of human TRIMSa. fused
directly to about
full-length human cyclophilin A. In another embodiment. the TRIM5-cyclophilin
fusion protein
comprises amino acids I to about 322 of human TT iM5cm fused directly to about
full-length
human cyclophilin A. In still another embodimnent, the TRIM5-cyclophilin
fusion protein
comprises amino acids I to about 331 of human TI I '15a fused directly to
about full-length
human cyclophilin A. Other suitable proteins that inhibit HIV replication that
can be encoded by
the second nucleic acid sequence include, but are not limited to, cyclophilin,
E3 ubiquitin,
APOBEC3 _i, and bone marrow strornal cell antigen 2 (1IST-2).
19

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0063] The nucleic acid sequences of the present invention further include
nucleic acid
sequences that encode conservative variants or functional equivalents of the
proteins herein
described. As used herein, a conservative variant refers to alterations in the
amino acid sequence
that do not adversely affect the biological functions of the protein, A_
substitution, insertion or
deletion is said to adversely affect the protein when the altered sequence
prevents or disrupts a
biological function associated with the protein. For example, the overall
charge, structure or
hydrophobic/hydrophilic properties of the protein may be altered without
adversely affecting a
biological activity. Accordingly, the amino acid sequence can be altered, for
example to render
the protein more hydrophobic or hydrophilic, without adversely affecting the
biological activities
of the protein.
[0064] Ordinarily, the conservative substitution variants and functional
equivalents of the
proteins, will have an amino acid sequence identity to the disclosed sequences
SEQ ID NOs: 2
and 4 of at least about 55"f%, at least about 65%, at least about 75"o, at
least about 80%, at least
about 85%, at least about 90%. at least about 95 %3, or at least about 96" to
99%. Identity or
homology with respect to such sequences is defined herein as the percentage of
amino acid
residues in the candidate sequence that are identical with the known peptides,
after aligning the
sequences and introducing gaps, if necessary, to achieve the maximum percent
homology, and
not considering any conservative substitutions as part of the sequence
identity. N-terminal, C-
terminal or internal extensions, deletions, or insertions into the peptide
sequence shall not be
construed as affecting homology.
[0065] Thus, the nucleic acid sequences of the expression vectors of the
present invention can
encode conservative variants or functional equivalents of the protein
sequences described herein.
Contemplated variants further include those containing predetermined mutations
by, e.g.,
homologous recombination. site-directed. or IICR mutagenesis, and. the
corresponding proteins of
other animal species, including but not limited to rabbit, rat, porcine,
bovine, ovine, equine and
non-human primate species.

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0066] In some embodiments of the expression vectors of the invention, the
first nucleic acid
sequence encoding an inhibitor of an HIV co-receptor is operably linked to a
first promoter and
the second nucleic acid sequence encoding a protein that inhibits HIV fusion
to a target cell or
HIV replication is operably linked to a second promoter. In certain
embodiments of the
invention in which the expression vector comprises three nucleic acid
sequences, each of the
three nucleic acid sequences can be operably linked to a separate promoter.
For instance, in one
embodiment, the first nucleic acid sequence encoding an inhibitor of an HIV co-
receptor is
operably linked to a first promoter, the second nucleic acid sequence encoding
a protein that
inhibits HIV fusion to a target cell is linked to a second promoter, and the
third nucleic acid
sequence encoding a protein that inhibits HIV replication is operably linked
to a third promoter.
In other embodiments, two of the three nucleic acid sequences are transcribed
from a single
promoter (i.e. the first and second nucleic acid sequences or the second and
third nucleic acid
sequences). In still other embodiments, all three nucleic acid sequences are
transcribed from a
single promoter. All three promoters can be the same or different fromn one
another.
100671 The phrase "operably linked" or "under transcriptional control" as used
herein means that
the promoter is in the correct location and orientation in relation to a
nucleic acid sequence to
control the initiation of transcription by RNA polynrerase and expression of
the nucleic acid. The
promoters selected preferably lack promoter exclusion, thereby avoiding one
promoter switching
off the other promoter(s). The first, second, and third promoters can be RNA
polymerase I (,pot
I), polyrnerase 11 (pot II), or polyrnerase III (pol 111) promoters. The
promoters may be
constitutive promoters or inducible promoters. Inducible promoters are known
in the art and can
include the tetracycline promoter, metallothionein II A promoter, heat shock
promoter,
steroid/thyroid hormone/retinoic acid response elements, the adenovirus late
promoter, and. the
inducible mouse mammary tumor virus LTR. In one embodiment, the promoter
contains at least
a portion of an HIV LTR (e.g. , TAR) and is inducible by HIV infection. In
certain embodiments,
the first promoter is a RNA pot III promoter. RNA pot III promoters suitable
for use in the
expression vectors of the invention include, but are not limited., to human
U6, mouse U6, and
human H I . In one embodiment, the first promoter is a HI RNA pot III
promoter. In other
embodiments, the second promoter is a RNA pol 11 promoter. In one particular
embodiment, the
second promoter is a IJbiquitinC pol 11 promoter. The second promoter, in some
embodiments,
21

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
can be a tissue-specific promoter. For instance, suitable tissue-specific
promoters include
macrophage-specifie promoters (e.g., MPG-I and the like) and 'f-cell promoters
(e.g., CD 4 and
the like). In one embodiment, the third promoter is a RIN '4 pol II promoter.
In another
embodiment, the third. promoter is a UhiquitinC pol II promoter. The third
promoter can, in
some embodiments, be a tissue specific promoter. The first, second, and third
promoters can be
a combination of any of the promoters described herein. In certain
embodiments, RNA pol III
promoters are preferred where the nucleic acid sequence encodes an inhibitory
RN molecule,
such as an siR :A or shRITA, In other embodiments, RNA pol II promoters are
preferred where
the nucleic acid sequence encodes a protein.
[0068] In embodiments in which the inhibitor of the I-TILT co-receptor is a
siRNA molecule, more
than one promoter ina-v be used to generate the siRNA molecule. For instance,
in one
embodiment, the expression vector comprises one nucleic acid molecule encoding
the sense
strand of the siRNA molecule and another nucleic acid molecule encoding the
antisense strand of
the silo.] A molecule such that the si RNA duplex is formed following
expression of the two
nucleic acids. In such embodiments, the expression vector can comprise a first
Pol III promoter
operably linked to the first nucleic acid encoding the sense strand and a
second fool III promoter
operably linked to the second nucleic acid encoding the antisense strand. In
another embodiment,
the expression vector comprises a first RNA Pol III promoter operably linked
to the first nucleic
acid sequence encoding the siRNA molecule targeting the 1-11 `o% co-receptor,
and a second RNA
Pol III promoter operably linked to the same first nucleic acid sequence in
the opposite direction,
such that expression of the first nucleic acid sequence from the first RNA Pol
I l promoter results
in a synthesis of the sense strand of the siRINA molecule and expression of
the first nucleic acid
sequence from the second RNA Pol III promoter results in synthesis of the
antisense strand of the
siRNA molecule. Following expression of the first nuclei: acid sequence from
the two different
promoters, the sense and antisense strands hybridize to form the duplex silA,
[0069] In one embodiment, the first nucleic acid sequence and the second
nucleic acid sequence
are transcribed from a single promoter. For instance, the first nucleic acid
sequence and the
second nucleic acid sequence are operably linked to a promoter such that a
single transcript is
generated, In another embodiment, an internal ribosome entry site (IRES) is
located upstream of
22

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
the second nucleic acid sequence and downstream of the first nucleic acid
sequence. In other
embodiments in which the expression vector comprises three nucleic acid
sequences, two of the
three nucleic acid sequences are transcribed from a single promoter (i.e. the
first and second
nucleic acid sequences or the second and third nucleic acid sequences. In
still other
embodiments, all three nucleic acid sequences are transcribed from a single
promoter. One or
more TREE elements can be present upstream of the second and/or third nucleic
acid sequences.
For instance, in one embodiment, the first, second, and third nucleic acid
sequences can be
operably linked to a single promoter and a first IRES element can be
positioned between the first
and second nucleic acid sequence and a second IRES element can be positioned
between the
second and third nucleic acid sequence. IRES elements enable efficient
translation of
polycistronic messages. Any IRES element known in the art can be used in the
expression
constructs of the invention.
100701 In certain embodiments, the first and second nucleic acid sequences are
expressed in
different ratios such that expression of the HIV co-receptor inhibitor will be
higher than that of
the HIV replication or fusion inhibitor. For instance, the ratio of expression
of the first nucleic
acid sequence to the second nucleic acid sequence can be from about 2:1 to
greater than about
10:1, preferably from about 5:1 to about 10:1, more preferably from about 2:1
to about 5:1. In
one embodiment, the ratio of expression of the first nucleic acid sequence to
the second nucleic
acid sequence is about 2:1. In embodiments in which the expression vector
comprises three
nucleic acid sequences, the ratio of expression of the first, second, and
third nucleic acid
sequences can be manipulated such that the expression of HIS' co-receptor
inhibitors will be
higher than that of the HIV replication and. fusion inhibitors. For instance,
in embodiments in
which the first nucleic acid sequence encodes an inhibitor of an HIV co-
receptor E e.g. CCR5 or
CXCR44lo the second nucleic acid sequence encodes a fusion inhibitor, and the
third nucleic acid
sequence encodes a replication inhibitor, the ratio of expression of the
first, second, and. third,
nucleic acid sequences can be from about 2:1:1 to about 10:1: 1, from about 5:
1:1 to about 10:1: 1,
or from about 2:1:1 to about 5:1:1.
[0071] Generation of the expression vectors described herein can be
accomplished using any
suitable genetic engineering techniques well known in the art, including,
without limitation, the
13

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
standard techniques of PCR, oligonucleotide synthesis, restriction
endonuclease digestion,
ligation, transformation, plasiuid purification, and DNA sequencing, for
example as described in
Sambrook et al. (Molecular Cloning--A Laboratory Manual (3rd Ed.), Vol. 1-3,
Cold Spring
Harbor Laboratory, Cold Spring Harbor, N.Y., (2000)), Ausubel et al. (Current
Protocols in
Molecular Biology, Greene Publishing; Associates, Inc. and John Wiley & Sons,
inc.), Coffin et
a1. (Retroviruses. Cold Spring Harbor Laboratory Press, -N.Y. (1997)) and "RNA
Viruses: A
Practical Approach" (Alan I. Cann, Ed., Oxford University Press, (2000)).
[0072] In one embodiment, the expression vector is an FG12 vector, and more
preferably an
F a 1 IF lentiviral vector (See Example 1). In another enmibodirnent, the
second nucleic acid
sequence is cloned into two restriction sites (e.g., Bainlll and EcoRI of the
PG]-1 F vector). In
yet another embodirnerit, the first nucleic acid sequence is inserted between
two restriction sites
(e.g., Xbal/Xhol sites of the F(aI IF vector). In certain embodiments, the
viral expression vector
further comprises at least one further nucleic acid molecule capable of
inhibiting Ii IV infection,
selected from a shRlsiA or siRlsiA, an aritisense molecule, a ribozynie or an
aptamer targeted to a
1ilY viral sequence or host sequence. In other embodiments, the viral
expression vector further
comprises one or more protein-encoding nucleic acid sequences as described
herein. For
instance, in one embodiment, the viral expression vector further comprises one
or more nucleic
acid sequences encoding protein inhibitors of HIV viral fusion or IV
replication.
100731 The novel expression vectors of the invention confer resistance to
infection by more than
one 111V variant when expressed in host cells. In one embodiment, the novel
expression vectors,
when expressed in host cells, confer resistance to infection by R5- and X4-
tropic strains of HIV.
In some embodiments, when expressed in host cells, the expression vectors
confer resistance to
infection by strains of HIV that are resistant to HAART or Maraviroc
therapies.
[0074] The present invention also includes a method of producing a viral
expression vector that
is capable of inhibiting binding of HIV to the cell and preventing HIV fusion
to the cell or HIV
replication when expressed in a host cell. In one embodiment, the method
comprises
synthesizing a eDNA of a gene which expresses a protein capable of preventing
24

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
HIV fusion into a cell or HIV replication; cloning the synthesized cDNA into a
restriction site in
a viral vector; and inserting an expression unit capable of downregulating
expression of an HIV
co-receptor into a restriction site in the vector. The cDNA can be from any
gene which
expresses any of the protein fusion or replication inhibitors described
herein. For instance, in
one embodiment, the cD- :A is a C46 eDN:A. In another embodiment, the eDr :A
is a TRIM5a,
eD:N A or a cyclophilin fusion thereof. The expression unit capable of
downregulating
expression of a HIV co-receptor can be any of the inhibitory RNA molecules
described herein,
such as silA, shR'-A, or antisense targeting the co-receptor. In one
embodiment, the
expression unit is a shR A targeting CCR5. In one particular embodiment, the
shR A targeting
CCR5 has a sequence of SECS III NO: 1.
[0075] The viral vector can be a retroviral vector. In certain embodiments,
the viral vector is a
lentiviral vector. such as the FG I IF lentiviral vector. In some embodiments,
the cDNA of a gene
encoding a protein fusion or replication inhibitor is cloned into restriction
sites BamI-II and
TcoRI of an FEl I IF vector. In other embodiments, the expression unit capable
of
downregulating expression of an HIS' co-receptor is inserted between XbaUXhol
restriction sites
of the FGI IF vector. Other lentiviral vectors and restriction sites suitable
for use in the method
are known to those of ordinary skill in the art.
[0076] The present invention also provides a host cell comprising the novel
expression vectors of
the invention. A "host cell" or "target cell" means a cell that is to be
transformed using the
methods and expression vectors of the invention. In some embodiments, the host
cells are
mammalian cells in which the expression vector can be expressed. Suitable
mammalian host
cells include, but are not limited to, human cells, r-marine cells, non-human
primate cells (e.g.
rhesus monkey cells), human progenitor cells or stern cells, 293 cells, I-
IeI:a cells, DI 7 cells,
,\MM-DC'K _ cells, BI-II cells, and (12Th cells. In certain embodiments, the
host cell comprising an
expression vector of the invention is a hematopoietic cell, such as
hematopoietic progenitor/stem
cell (I I CID34-positive henaatopoietic progenitor/stem cell (I-IPSC;)), a
monoeyte, a
macrophage, a peripheral blood mononuclear cell, a CD4 - T lymphocyte, a CD8+
I
lymphocyte, or a dendritic cell. In some embodiments, the host cell is a CCR5+
heanatopoietic
cell. other embodiments, the host cell may be a host cell from a patient or
matched to a

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
patient. In certain embodiments, a host cell transduced with the expression
vectors of the
invention are resistant to infection by X4 or R5 -tropic HIV strains,
including H_<" RT resistant
strains.
[0077] Methods of delivering expression vectors and nucleic acids to cells are
known in the art
and can include, for example, viral infection, calcium phosphate co-
precipitation,
electroporation, microinjection, DEAE-dextran, lipofection, transtection
employing polyamine
transfection reagents, cell sonication, gene bombardment using high velocity r
icroprojectiles,
and receptor-mediated transfection.
[0078] The present invention also encompasses a pharmaceutical composition
comprising the
novel expression vectors of the invention. In one embodiment, the
pharmaceutical composition
comprises an effective amount of at least one of the expression vectors as
described herein and a
pharmaceutically acceptable carrier. For instance, in certain embodiments, the
pharmaceutical
composition comprises an effective amount of an expression vector and a
pharmaceutically
acceptable carrier, wherein said expression vector coraprises a first nucleic
acid sequence
encoding an inhibitor of an i-IIV co-receptor and a second nucleic acid
sequence encoding a
protein that inhibits HIS' fusion to a target cell or HIV replication, as
described herein.
100-1,91 The phrases "pharmaceutically acceptable"or "pharmacolo ically
acceptable" refer to
molecular entities and compositions that do not produce adverse, allergic, or
other untoward
reactions when administered to an animal or a human. As used herein,
"pharmaceutically
acceptable carrier" includes solvents, buffers, solutions, dispersion media,
coatings, antibacterial
and antifungal agents, isotonic and absorption delaying agents and the like
acceptable for use in
formulating pharmaceuticals, such as pharmaceuticals suitable for
administration to humans.
The use of such media and agents for pharmaceutically active substances is
well known in the
art. Except insofar as any conventional media or agent is incompatible with
the expression
vectors of the present invention, its use in therapeutic compositions is
contemplated. The
pharmaceutical compositions of the invention may be formulated for
administration by various
routes of administration including, but not limited. to, oral" nasal, buccal,
intradermal,
subcutaneous, intramuscular, intraperitoneal, or intravenous injection. In
some embodiments,
26

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
the pharmaceutical compositions may be formulated as suppositories for rectal
administration.
Supplementary active ingredients also can be incorporated into the
compositions, provided they
do not inactivate the vectors or polynucleotides of the compositions.
[0080] The pharmaceutical compositions of the present invention may include
classic
pharmaceutical preparations. By way of illustration, solutions of the active
compounds as free
base or pharmacologically acceptable salts can he prepared in water suitably
mixed with a
surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared
in glycerol, liquid
polyethylene glycols, and mixtures thereof and in oils. Under ordinary
conditions of storage and
use, these preparations generally contain a preservative to prevent the growth
of microorganisms.
100811 The pharmaceutical forms suitable for injectable use include, for
example, sterile aqueous
solutions or dispersions and sterile powders for the extemporaneous
preparation of sterile
injectable solutions or dispersions. Generally, these preparations are sterile
and fluid to the
extent that easy injectability exists. Preparations should be stable under the
conditions of
manufacture and storage and should be preserved against the contaminating
action of
microorganisms, such as bacteria and #urigi. Appropriate solvents or
dispersion media may
contain, for example, water, ethanol, polyol (for example, glycerol, propylene
glycol, and liquid
polyethylene glycol, and the like), suitable mixtures thereof, and vegetable
oils. The proper
fluidity can be maintained, for example, by the use of a coating-, such as
lecithin, by the
maintenance of the required particle size in the case of dispersion and by the
use of surfactants.
The prevention of the action of microorganisms can be brought about by various
antibacterial
and antifurigal agents, for example, parabens, ehlorobutanol, phenol, sorbic
acid. thimerosal, and
the like. In many cases, it will be preferable to include isotonic agents, for
example, sugars or
sodium chloride. Prolonged absorption of the injectable compositions can be
brought about by
the use in the compositions of agents delaying absorption, for example,
aluminum monostearate
and gelatin.
[00821 Sterile injectable solutions may be prepared by incorporating the
active compounds in an
a 11
appropriate amount into a solvent along with any other ingredients (for
example as enumerated
above) as desired, followed by filtered sterilization. Generally, dispersions
are prepared by
27

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
incorporating the various sterilized active ingredients into a sterile vehicle
which contains the
basic dispersion medium and the desired other ingredients, e.g., as enumerated
above. In the
case of sterile powders for the preparation of sterile injectable solutions,
the preferred methods of
preparation include vacuum-drying and freeze-drying techniques which yield. a
powder of the
active ingredient(s) plus any additional desired ingredient from a previously
sterile-filtered
solution thereof.
[00831 The compositions of the present invention generally may be formulated
in a neutral or
salt form. Pharmaceutically-acceptable salts include, for example, acid
addition salts (formed
with the free amino groups of the protein) derived from inorganic acids (e.g.,
hydrochloric or
phosphoric acids), or from organic acids (e g., acetic, oxalic, tartaric,
mandelic, and the like)).
Salts formed with the free carboxyl groups of the protein can also be derived
from inorganic
bases (e.g., sodium, potassium, ammonium, calciurm, or ferric hydroxides) or
from organic bases
(e.g., isopropylannine, trimethylannine, histidirne, procairie and the like).
100841 The present invention also includes a method of treating or preventing
HIV infection in a
patient in need thereof. As used herein, "patient" or "subject" may encompass
any vertebrate
including but not limited to humans and mamrmals. Ilowever, advantageously,
the patient or
subject is a mammal such as a human or non-human primate, or a mammal such as
a
domesticated mammal, e.g., dog, cat, horse, and the like, or production
mammal, e.g., cow,
sheep, pig, and the like. Where the patient is other than human, the invention
provides a method
of treating or preventing an HIV-related infection in the patient (e.g.,
infection by Sly, Fl V, or
BIV). In a preferred embodiment, the patient is a human.
[00851 In one embodiment, the method comprises administering a pharmaceutical
composition
comprising an expression vector of the invention as described herein, For
instance, in some
embodiments, the method comprises administering to the patient a
pharmaceutical composition
comprising an expression vector wherein said. expression vector comprises a
first nucleic acid
sequence encoding a shRNA targeting CCR5 (or CXCR4II and a second nucleic acid
sequence
encoding a C46 protein, and optionally wherein said first nucleic acid.
sequence is operably
linked to a first promoter and said second nucleic acid sequence is operably
linked to a second
28

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
promoter as described. In other embodiments, the method comprises
administering to the patient
a pharmaceutical composition comprising an expression vector wherein said
expression vector
comprises a first nucleic acid sequence encoding a shRNA targeting CCR5 (or
CXCR4) and. a
second nucleic acid sequence encoding a TRIM5(k protein or derivative or
fusion thereof, and
optionally wherein said first nucleic acid sequence is operably linked to a
first promoter and said
second nucleic acid sequence is operably linked to a second promoter as
described. In still other
embodiments, the method comprises administering to the patient a
pharmaceutical composition
comprising an expression vector wherein said expression vector comprises a
first nucleic acid
sequence encoding an inhibitor of an HIV co-receptor (e.g., sh~"NT A to CCR5
or CXCR4), a
second nucleic acid sequence encoding a fusion inhibitor (e.g., C46), and a
third nucleic acid
sequence encoding an inhibitor of HIV replication (e.g., T M5 , protein or a
derivative or
fusion thereof), optionally wherein said first, second, and third nucleic acid
sequences are
operably linked to first, second, and third promoters as described herein. In
another embodiment,
the method comprises administering to the patient a pharmaceutical composition
comprising an
expression vector wherein said expression vector comprises a first nucleic
acid sequence
encoding a first inhibitor of an HIV co-receptor (e.g., shRNA to CCR5), a
second nucleic acid
sequence encoding a second inhibitor of an 1-1 V co-receptor (e.g., shRNA to C
XC R4), and a
third nucleic acid sequence encoding an inhibitor of HIV viral fusion to a
target cell or HIY
replication, optionally wherein said first, second, and third nucleic acid
sequences are operably
linked to first, second, and third promoters as described herein.
[00861 In certain embodiments, the patient to whom the pharmaceutical
composition is
administered is a patient at risk of infection by R5 and X4 tropic strains of
HIV, including
HART resistant strains, and such risk is ameliorated following administration
of the
composition. In some embodiments, the patient is HIS' negative. In other
embodiments, the
patient (e.g., human) may be lIl positive and naive to highly active
antiretroviral therapy
(I HART) -that is the human patient has never received HART, which includes
combinations
of nucleoside analogue reverse transcriptase inhibitors, protease inhibitors,
and non-nucleoside
reverse transcriptase inhibitors. In some embodiments, the patient is
receiving a. HAAI: T
regimen. In still other embodiments, the patient is failing or has failed on a
HAART regimen
(i.e. IIAART is ineffective in reducing viral load due to resistance). Thus,
in certain
29

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
embodiments, the expression vector is introduced directly to the patient
either prophylatically for
a patient who is HIV negative or to treat a patient who is HIV positive.
[00871 The expression vectors of the compositions can be modified such that
they are
specifically localized to particular cell types, such as immune cells. By way
of example, the
expression vector may be combined with a receptor-mediated gene targeting
vehicle, wherein
said targeting vehicle comprises a cell-receptor-specific ligand and a DNA-
binding agent.
Alternatively, a cell receptor-specific ligand can be attached to a liposorne
comprising the
expression vector. The cell-receptor-specific ligands can be chosen depending
on the cell types
of interest. For instance, in some embodiments, the expression vector may be
localized to
CD34 cells by employing a ligaand that binds to the CD34 cell surface marker,
One of skill in
the art can choose appropriate ligands to target specific cell types, such as
immune cells (e.g., a
nionocyte/ acrophage, a peripheral blood mononuclear cell, a CD4-f T
lymphocyte, a CD$-f- T
lymphocyte, or a dendritic cell). In certain embodiments in which the
expression vector is a viral
vector, the viral vector can be packaged in viral particles having a
particular tropism for certain
cell types. For example, in one enibodirnent, the viral vector is packaged in
II IV retroviral
particles thereby allowing the recombinant retirovirus to infect CD4 T cells
and macrophages.
10088 Administration to a patient of the pharmaceutical compositions according
to the present
invention may be via any common route so long as the target tissue is
available via that route.
This includes oral, nasal, or buccal. Alternatively, administration may be by
intradermal,
subcutaneous, intramuscular,, intraperitoneal or intravenous injection. In one
embodiment, the
pharmaceutical composition maybe administered. rectally (e.g., with a
suppository). Upon
formulation, solutions are preferably administered in a manner compatible with
the dosage
formulation and in such amount as is therapeutically effective. The
formulations may easily be
administered. in a variety of dosage forms such as injectable solutions, drug
release capsules and
the like. For parenteral administration in an aqueous solution, for example,
the solution
generally is suitably buffered and. the liquid diluent first rendered isotonic
for example with
sufficient saline or glucose. Such aqueous solutions may be used., for
example, for intravenous,
intramuscular, subcutaneous and intraperitoneal administration. Preferably,
sterile aqueous
media are employed as is known to those of skill in the art, particularly in
light of the present

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
disclosure. By way of illustration, a single dose may be dissolved in 1 ml of
isotonic NaC l
solution and. either added to 1000 ml of hypodermoclysis fluid or injected at
the proposed site of
infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th
Edition, pages 1035-
1038 and 1570-1580). Some variation in dosage may occur depending on the stage
of HIV
infection in the patient being treated. The person responsible for
administration will, in any
event, determine the appropriate dose for the individual patient. Moreover,
for human
administration, preparations should meet sterility, py-rogenicit:y, general
safety and purity
standards as required by FDA Office of Biologics standards.
[00891 In another embodiment, the present invention provides a method of
treating or preventing
HIS' infection in a patient by administering to the patient HIV-resistant
hematopoietic cells
produced by trarnsducing the cells with an expression vector of the invention.
For example, in
one embodiment, the method comprises transducing hematopoietic cells ex vivo
with an
expression vector described herein, and infusing the transduced cells into the
patient. One or
more infusions of the transduced cells can be administered to the patient. In
some embodiments,
the patient receives multiple infusions of the trarnsduced cells over a
periodic interval, such as
weekly, biweekly, monthly, quarterly, or annually. In one embodiment, the
patient receives an
infusion of the transduced cells every two weeks. llematopoietic cells
suitable for use in the
method include, but are not limited to, hcmatopoietic progenitor/stem cells
6HPSC), monocytes,
macrophages, peripheral blood mononuclear cells, CD4+ T lymphocytes, C D8 - T
lymphocytes,
and dendritic cells. In one embodiment, the hemnatopoietic cells used in the
method are CD4+ T
lymphocytes, CD8+ T lymphocytes, or rnonocyte/macrophages. In a preferred
embodiment, the
hematopoietic cells used in the method are HPSC, As used herein, transduced.
hematopoietic
cells include the transduced. cells themselves as well as cells derived from
the transduced cells
(e g., cells generated from transduced HPSQ.
[00901 Thus, in one particular embodiment, the present invention provides a
method of treating
or preventing HIV infection in a patient by reconstituting the immune system
with HIV-resistant
cells generated from transduced HPSC:. For instance. in one embodiment, the
method. comprises
transducing HPSC with an expression vector as described. herein and.
transplanting said
transduced HPSC: in the patient, wherein said transplanted cells generate
granulocytes,
31

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
monocyte/macrophages, and lymphocytes that are resistant to HIV infection. The
granulocytes,
monocyte/macrophages, and lymphocytes are resistant to infection by R5 and X4
tropic strains
of HIV. In some embodiments, the granulocytes, monocvte/macrophages, and
lymphocytes are
resistant to infection by HIV strains that are resistant to H.'\-ART. The
patient can be HIV
negative or IIIV positive. In one embodiment, the human patient is naive to
highly active
antiretroviral therapy (H. ART ) In another embodiment, the patient is
receiving a HAART
regimen. In yet another embodiment, the patient is failing or has failed on a
H. TAT regimen.
[0091] The hematopoietic cells (e.g. HPSC, CD4+ T lymphocytes, CD8+ T
lymphocytes, and/or
monocyte/macrophages) to he transdctced with an expression vector of the
invention can be
allogeneic or autologous. "Allogeneic cells" refer to cells obtained from
different individuals of
the same species. As used herein, the phrase "autologous cells'"' refers to
cells isolated from a
patient that are subsequently reimplanted or injected into the same patient.
Thus, an autologous
transplantation is one in which the donor and recipient are the same patient.
In certain
embodiments, the hernatopoietic cells are autologous TIPSC. The HPSC are
preferablyr CCD34-
positive and can be isolated from the patient's bone marrow or peripheral
blood. Methods for
such purification are known to those in the art (see, for example, U .S.
Patent Nos.: 4,965,204,
4,714.680, 5,061.620, 5,643,741, 5,577,136, 5,716,827, 5,750,397, and
5,759,793). For instance,
one method for purifying such C D34-positive steer cells involves
centrifugation of peripheral
blood samples to separate mononuclear cells and granulocytes, followed by
fluorescence
activated cell sorting (FACS) to select CD34-+ cells. In one embodiment, the
cells are enriched
for CD34 cells through a magnetic separation technology such as that available
from Miltenyi
Biotec and that has been previously described (Kogler et al. (1998) Bone
Marrow 'Transplant.,
Vol. 21:233-241, Pasino e al. (2000) Br. J. Haematol., Vol. 108: 793-800).
CD34-positive cells
may be mobilized from the marrow into the blood prior to collection by
injecting the patient with
one or more cytokines known to mobilize hematopoictic stem cells, such as
granulocyte colony
stimulating factor, tgranulocyte-macrophagc stimulating factor, and stem cell
factor.
[0092] The isolated. CD34-positive HPSC (and/or other hematopoietic cell
described. herein) is
preferably transduced with an expression vector of the invention. F'or
example. in one
embodiment the expression vector comprises a fast nucleic acid sequence
encoding an inhibitor
32

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
of an HIV co-receptor and a second nucleic acid sequence encoding a protein
that inhibits HIV
fusion to a target cell or HIV replication, optionally wherein said first
nucleic acid. sequence is
operably linked to a first promoter and said second nucleic acid. sequence is
operably linked to a
second. promoter. In another embodiment, the expression vector comprises a
first nucleic acid
sequence encoding an inhibitor of an I-TIV co-receptor, a second nucleic acid
sequence encoding
a fusion inhibitor, and a third nucleic acid sequence encoding an inhibitor of
HIV replication,
optionally wherein said first, second, and third nucleic acid sequences are
operably linked to
first, second, and third promoters. In still another embodiment, the
expression vector comprises a
first nucleic acid sequence encoding a first inhibitor of an HIV co-receptor,
a second nucleic acid
sequence encoding a second inhibitor of an HIV co-receptor, and a third
nucleic acid sequence
encoding an inhibitor of HTV viral fusion to a target cell or 11V replication,
optionally wherein
said first, second, and third nucleic acid sequences are operably linked to
first, second, and third
promoters.
[00931 In one embodiment, the first nucleic acid sequence (or second nucleic
acid sequence in
embodiments in which the expression vector comprises three nucleic acid
sequences) encodes a
siRNA or shRNA having a double-stranded region, wherein the double-stranded
region
comprises a sequence that is substantially identical and complementary to a
sequence of C;'C;'RS.
In another embodiment, the first nucleic acid sequence encodes a shRNA
targeting CCR5 that
has a sequence of SEQ ID No : 1. In another embodiment, the first nucleic acid
sequence
encodes a siRNA or shRNA having a double-stranded region, wherein the double-
stranded
region comprises a sequence that is substantially identical and complementary
to a sequence of
CXC R4. In particular embodiments, the transduced hematopoietic cells (e.g.
HPSC, C D4-1 T
lymphocytes, CD8+ I lymphocytes, and/or monocyte/macrophages) or cells
generated from
them express reduced levels of a HIV co-receptor (e.g. CCR5 or CXCR4) protein
as compared to
non-transduced hematopoietic cells. For instance the transduced hematopoietic
cells or cells
generated from them may express 30%, 40%, 50%, 60%, 65%, 70%, 15%, 80%, 85%,
90%, or
95% less HIV co-receptor protein as compared to non-transduced. hematopoietic
cells. In other
embodiments, the hematopoietic cells are transduced with an expression vector
of the invention
in which the second nucleic acid sequence (or third nucleic acid. sequence in
embodiments in
which the expression rector comprises three nucleic acid sequences) encodes a
TRTM5u , protein
33

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
or derivative or fusion thereof, such as human TRIM' a, rhesus TRIMS a,
chimeric TRIM' (X,
or a human TRT MM_S-cyclophilin fusion protein. In still other
enrbodirrrerrts, the hernatopoietic
cells are transduced with an expression vector of the invention in which the
second nucleic acid
sequence or third nucleic acid sequence in embodiments in which the expression
vector
comprises three nucleic acid sequences) encodes a C46 protein. In such
embodiments, the
transduced hernatopoietic cells or cells generated from there express
increased levels of the
protein (e.g. C46 or TRINI5(x or derivative or fusion thereof) as compared to
non-transduced
, that is 20 Q, 30%, 40 %, 50 %3, 60"%%, 70%. S0 Q, 90%, 1 OWN,, 150,'.!
hernatopoietic cells, or
greater than 200`/() more of the encoded protein as compared to non-transduced
hematopoietic
cells.
100941 Following transduction of the hernatopoietic cells (e.g., HPSC, CD4 - T
lymphocytes,
CD8+ I lymphocytes, or monocyte/macrophages) with an expression vector of the
invention, the
transduced cells are reintroduced or transplanted back into the patient. The
transduced cells can
be injected parenterally into the patient, or reintroduced by any other route
brown in the art. In
one embodiment, the transduced hernatopoietic cells are injected intravenously
into the patient.
Preferably, an effective dose of transduced hematopoietic cells is
administered to the patient. An
effective dose" is an amount sufficient to effect a beneficial or desired
clinical result and can
depend on the type of hematopoietic cell used. In one embodiment, the
hematopoietic cell is a
HPSC and an effective dose is an amount that is sufficient to at least
partially reconstitute the
immune system with HIV-resistant cells. Said dose could be administered in one
or more
administrations and may be from about 0.5 x 106 HPSC per kg patient weight to
about 1 x 109
HPSC per kg patient weight. In another embodiment, the hematopoietic cell is a
CD4+ T
lymphocyte, a CD8+ T lymphocyte, or a rnonocyte/macrophage and an effective
dose may be
from about I x 109 cells per patient to I x 1011 cells per patient. However,
the precise
determination of what would be considered an effective dose may be based on
factors individual
to each patient, including their size, age, severity of HIV infection (e.g.
viral titer), and amount
of tirne since contraction of the virus. One skilled in the art, specifically
a physician, would be
able to determine the number of transduced hernatopoietic cells which would
constitute an
effective dose without being subjected to undue experimentation.
34

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0095] Without being bound by theory, Applicants believe that a successful
stem cell therapy for
HIV disease includes selection for transduced, engrafted cells. In one aspect
of the invention,
detailed kinetic studies on HI\' infected individuals demonstrate that HIV- 1
kills and the body
replenishes approximately 109 to 1010 CD4+ T-cells each day. This represents a
turnover of 0.5%%
to 5% of the total CD4+ T-cell population each day resulting in an estimated
turnover of the
entire CD4+ T-cell population approximately every 2 weeks. Therefore, even in
healthy
untreated HIV infected individuals, a stable CD4+ T-cell count masks massive
ongoing death
and replenishment of T-cells. T-cells are replaced from two sources-expansion
of existing
peripheral T-cells, and production of new naive T-cells derived from the
thymus, in a manner
similar to that seen in generation of new T-cells following HPSC transplant.
The present
invention provides a method to reconstitute with gene-modified HPSC that
provide a continual
source of protected T-cells and monocyte/macrophages. These cells are likely
to be selected for
in the face of massive 111V T-cell depletion.
[0096] The concept of utilizing selective pressures involving T-cell death and
regeneration to
select for gene-transduced cells is based upon a solid foundation of knowledge
in hematopoietic
and lymphoid differentiation. The concept has also been tested successfully in
the gene therapy
clinical studies for X-linked SC:ID and ADA-SCID where gene containing T-cells
derived from
transplanted HPSC are similarly selected resulting in repopulation with
genetically modified T
cells.
10097 A recent case study provides support that reconstitution of an immune
system with cells
protected from HIV- I infection can result in selection for the protected
cells, substantial
attenuation of HIV-1 replication and a favorable clinical course. An HIV-I
positive individual
with concurrent AML was treated by transplant of allogeneic HPSC specifically
chosen from a
C,C,R5A32) . homozygous donor. Remarkably, the CCR5A3". donor cells completely
replaced. the
recipient cells within a rapid 6() days and the patient has remained
undetectable for HIV- I for
more than 200 days in the absence of anti-retroviral therapy.

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[0098] The invention will now be illustrated in greater detail by reference to
the specific
embodiments described in the following examples. The examples are intended to
be purely
illustrative of the invention and are not intended to limit its scope in any
way.
EXAMPLES
Example 1. Construction of Dual Vector Containing sh lA against CCR5 and C46
Fusion
Inhibitor (shy/ C46 Dual Vector) and Control Vectors with Single or no
Therapeutic Inserts
A. Vector Plasmid Constructs
[0099] A variety of constructs were designed and engineered in the DNA form as
plasrnids, The
constructs are summarized in Table l and illustrated in Figures 1-4. All of
these constructs give
rise to lentiviral vectors upon transfection into packaging cell lines (see
section B below).
Table 1. Description of Vector Plasmid Constructs
PlasmÃd Construct
Full Name Short Nam Description
e
pF 1I F-U- GFP Control Control single lentiviral vector (LV) containing
EGFP u iquitin prgmotÃ;r driving ECFP
--- -------------------- -----------------------------------------¾ -------
pF 12xH1NR5 shy Single LV containing H1 promoter driving
shRNA against CCR5
pFG1 I F-U6 C46 Single LV containing ubiquitin promoter driving
C46 C46
---- ----------------- ---------- --- -----------------------------------------
----------------------------------------------------------
pF'GI1 F-H1 A sh5/C46 Dual L\/ containing H1 promoter driving shRNA
R5-U-C46 against CCR5 and ubiquitin promoter driving
C46
pFG12-H1= sh5/GFP LV containing H1 promoter driving shRNA
R5 U EGFP (also referred against CCR5 and uÃb qu tin promoter driving
to as sh/EGFP) EGFP
1991001 The genetic engineering of these vectors was as follows.
1001011 The pFG 12 backbone lentiviral vector plasmid containing EGFP driven
by the ubiquitin
promoter (FFGl2-U-Et3FF) (labeled as "pFG12" in Figure 2) was derived from an
earlier
lentiviral vector FUGW (Lois et al. (2002) Science, Vol. 295: 868-872) as
described On el al.
(2003) Proc. Natl. Acad. Sci., Vol. 100: 183-188). 'I'o aid further insertions
into the backbone
36

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
vector, the plasmid backbone pFG I IF was produced by inserting multiple
cloning sites into
FG12, enabling production of pFGI IF-U-EGFFP (labeled as "pGI IF" in Figure
2).
[001021 A small hairpin RNA (shRNA) random library directed against human
chemokine co-
receptor 5 (huCCR5) under the control of an Hl promoter within a lentiviral
vector was
produced via enzymatic production of RNAi libraries from cDlr:As. The purified
DNA
fragments were digested with Bpn i, blunt-ended with Klenow fragment, digested
with Barnlll
and ligated to pBShHl _5 plasrnid DNA, which contains a human Hl RNA
polymerase Ill
promoter and 4T termination signal, The ligation mixture was introduced into
E. roll; and plated
overnight. Colonies were combined and plasnrid DNA prepared. shRNA expression
units
consisting of an Hl promoter, shRNA_ sequence and 4Ts termination signal were
excised from
the p13Sh111-5 plasrnid DNAs by XbaI and XholI digestion and insetted into
Xbal/Xholl sites of
the pFG12-I?-EGFP vector to produce 11I promoter driven shRNA against CCR5.
The best of
these constructs, shl005, was selected for further experimentation. The
plasinid construct
containing slrl 005 and ubiquitin 1?rc?r?router-dui ers l it P is termed 13F
_i 12-111 R5-I_T-EGFP
(Figure 3; An e/ al. (2007) Proc. Natl. Acad. Sci., Vol. 104 (32): 13110-
13115). The U-EGFP
cassette was removed from pFt 312-f-I1-R5-U-EG FP using restriction enzymes to
produce
pFt 312-1I1-R5 (Figure 3).
100103 The EGFI' gene was removed from pFG11F-U-EGFI' (pFG1 IF in Figure 4)
and
replaced with the C46 gene to produce pFG1IF-U-C'46 (Figure 4). The H1-R5
cassette was
excised from pFG12-1-I1-R54U-EGFI' using an Ndel / Xhol digest and inserted
into pFG1IF-U-
C46, which had also been digested with Ndel Xho?I, to produce pF'GI 1I;-HI-R5-
-C46 (Figure
4)o
B. Lentiviral Vector Production
[001041 All vesicular stornatitis virus (VSV)-G pseudotyped lentiviral vector
stocks were
produced by calcium phosphate-mediated transient transfection of HEIR-293 T
cells. HEIR-293 T
cells were routinely cultured in DMEM (GIBCO Invitrogen) and changed to
Iscove's modified
Dulbecco"s medium (IMDM) for transfection. All cultures contained. I0%'o FCS
(HyClone), 100
units of penicillin. and 100 ,tginil streptomycin. The cells were co-
transfecied with appropriate
377

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
amounts of vector plasmid, the HIV-1 lentiviral packaging constructs pRSV-Rev
and
pil Lg/p E, and. the VSV-G expression plasmid pCMV-VSV-G (Table 2). The
viruses were
collected from the culture supernatants on days 2 and 3 post-transfection and
concentrated.. The
concentrated virus stocks were titered. on HEIR-293 T cells based on GFP
expression, Titers for
the shR TA expression EGFP constructs were only slightly reduced compared with
the parental
EGFP vector. The plasmids used for production are shown diagrammatically in
Figure 5.
Table 2e Vectors for production of lentivirus
IasiÃd Description
-------------------------------------------------------------------------------
-------------------------------------------------------------------------------
---------------------------------------------------------
CMV-VSV-G VSVG envelope protein encoding plasmid
p Dt ! RRE Ga of RRE encoding lasmid
pRSV-Rev Rev encoding lasmid
Method 1: Lentivirus production by calciumn chloride transfection using non-
kit reagents
1. IIEK 293T eel Is seeded at 1.5x111' cells per Ti 75 flask in DMEM + 10% FBS
and
antibiotics the day prior to transfection.
2. On the day of transfection, medium changed to 25 mL IMDM with 111% FBS,
antibiotics and chloroquine(1l313 i of 10 mM).
3, DNA master mix prepared
a. pMDI,g /pRRE 10 ltg
b. pRS V-Rev 2.5 ~.tg
c. pC'MV-VSV-G13.2lug
d. vector (e.g. one of the constructs from Figs. 1-4) 10 l~,g
C. Water added to adjust total volume to 980 PL
4. 133 lrL 2M CaCl added, mixed and incubated on ice for 10 ruin.
5. 1110 t.tL 2 x El 13S (I g Elepes 1.6 M KCI)
g Naha, 0.72r_ril 0.25M Nazi-1P0
, I ml I
4
added drop by drop while shaking the tube by hand.
6. Incubated on ice for 20min.
T175 flask of cells tipped upside down, DNA mixture added to medium, flask
mixed 2-3 times and flask flipped right way up.
8. Culture incubated 6-8h
38

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
9. Medium removed and replaced with fresh 42inL IMDM + 10% FBS and
antibiotics.
10. 48h post transduction, medium harvested and filtered through 0,2.2. or
0.45 uM
filter and replaced with fresh 42anl IMDM + 10% FBS and antibiotics.
11, 72h post transduction, medium harvested and filtered through 0.22 or
0.45,uM
filter .
12, Both harvests are pooled
13. Virus containing medium (VCM,) concentrated by ultracentrifugation in SW28
or
SW32 tubes.
a. 33-38 ant, VCM loaded into tubes with sucrose cushion
b, Tubes centrifuged at 20,000 rpm at 4 C for 90 a-nine
c. Supernatant removed and 250-500 111, PBS or 1II3S added to pellet.
d. Store VCM overnight at 4 C: .
e. VC;M mixed by pipetting, aliquoted and stored at -70 C;.
Method 2: Lentivirus p production by calcium chloride transfectiorr using
Cloritech Calhhos Kit.
1. UEK 293T cells seeded at 2.1x1()' eells/"11225 in 30 mL IMDM + 10 %% FBS on
the day prior to transfection.
2. On the day of transfection, DNA master mix prepared in 15 FnL tubes:
a. pMDLg /pRRE 13 lL
h. pRSV-Rev 3.25 Leg
c. pCMV-VSV-G 4.16 .ug
d. vector (e.g. one of the constructs from Figs. 1-4) 13 ug
C. Water added to adjust total volume to 1500P1,
3. 186 ;uL. 2M C;aC1z added and mixed.
4. 1500 q[L 2 x FIBS added drop by drop while vortexing the tube.
5. Incubated at room temperature for 20 ruin.
6. 30 ml, I MI DM -;- 2%%% pf3S added to 50 mL tube.
7. DNA solution added to I-,\MM_DM in 50 L, tube.
8. Media aspirated from eel Is harvested the previous day.
9. DNA/'IM DM solution gently poured into flask so as riot to disturb cell
monol aver.
39

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
10. Flask gently rocked. from side to side to cover cells with mixture.
11. Culture inctibated. for 4. hours.
I2. Medium removed, cells rinsed. with PBS and. replaced with fresh 30 rnL
IMDM +
2%% FBS.
13, At 24 hours post transdcuc:tion, medium harvested and replaced with fresh
30 ]DI,
IMDM + 2% FBS.
14, harvested VCM filtered through 0.22 liM filter and stored at 4 C
overnight.
15. At 48 hours post transduction, medium harvested and filtered through 0.22
,op,M
filter .
16. Both VCM collections pooled and aliquoted for storage at 70 C.
17, If required, VCM concentrated using Vivaspin 20 (Sartorius) columns:
a. Vivaspin 20 MWCC? 100 000 prepared by adding 10 rn[_, 70% ethanol
h. Spun at 1000g for 10 min.
c. Remaining ethanol discarded and 15 rml: PBS added.
d. Spun at 1000g for 10 min.
C. Remainin PBS discarded and 1$ rnl: VCM 1 added.
f. Spun 1 OOOg for 30 min or until all VCM has passed through the column.
1001051 The VCM obtained by either method was used (diluted or concentrated)
to transduce
target cells (T cell lines, peripheral blood mononuclear cells (13BMC), CD34-f-
hematopoietic
progenitor stem cells (1113SC)) and the transduced cells were analyzed by flow
cytometry for
EGF13 expression, CCR5 expression (via CD195 antibody staining) and C46
expression (via 2F5
antibody staining).
Example 2. Transduction of Human Target I Cell Lines by sh5/C46 Dual Vector
[00106] The various lentiviral vectors described in Example I were used to
infect
CEM.NKR.CCR5 and Molt4/CCR5 cells (NIH AIDS Reagent Program) cells. 2 x ltd`
cells were
resuspended in ImL unconcentrated virus containing medium (VCM) with 10% FBS
and 8
p.g/rnL polybrene. Cultures were incubated at 3 7 C for 1.5 hours and a
further I mL of growth
media added (RPMI + 10% FBS), Cells were analyzed by FACS analysis 4 days post
transduoetio n t'6r C46 expression (by 2F5 antibody staining), CCR5 knockdown
(by CD 195

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
antibody staining), and GFP expression. Cells were kept in continuous culture
for up to 8 weeks
by passaging twice weekly.
[00107] Simultaneous expression of shy' (detected by CCR5 knockdown) and C46
in
transduced Cl M.NKR.CCRS and Molt4/CCR5 cells is shown in Figure 6 and Figure
7,
respectively. GFP expression was observed for the constructs containing EGFP
(panels 1,3 from
left to right), a reduction in CCR5 expression (e g. clown-modulation of CCR5
showing
expression of shRNA) was observed for the constructs containing shy (panels
2,3,5, from left to
right), and C46 expression (as measured by 2F5 antibody) was observed for the
constructs
containing C46 (panels 4,5, from left to right). Percentage positive cells are
shown in each flow
cytornetry% quadrant (Q1-Q4) for each group of cells transduced with the
indicated lentiviral
vectors at 4 and $ weeks in culture. Similar expression levels were seen at
weeks 4 and 8. Mean
fluorescence Intensity (MR) Values for Figure 6 are drown in Table .3 below,
while NMlF1 values
for Figure 7 are shown in Table 4 below.
Table 3. Mean Fluorescence Intensity Values for C.PM,NK,CCRS cells expressing
various
constructs
-------------------------------------- -------------------------------------- -
-------------------------------------- ------ ---------------------------------
------------------------------------- --------------------------------------
GPP control Shy Sh5/EGFP C46 Sh5!C46
4 weeks 70.2 33.8 10x5 78.2 11.5
------ --------------------------------- --------------------------------------
-- ---------------------------------------- ---------- ------------------------
------ ----------------------------------------
8 weeks 153.6 64.6 18.6 129.1 22.6
------------------------------ ----------
-------------------------------
Table 4. Mean Fluorescence Intensity Values for Molt4/CCR5 cells expressing
various
constructs
CPP control Sh5 Shy/ECPP C46 Sh5/C46
4 weeks 34.4 7.3 5.0 24.1 5,9
8 weeks 80.9 25.3 18,2 69.5 51.5
[00108] To determine if the transgenes caused any differences in the growth
parameters of the
cells, CEM.NKR.CCR5 cells, which each showed 100% expression of the transgenic
construct,
were seeded at 2 x 10' cells/n-A-, cultured for 4 days and. counted. Cells
were then seeded from
this population on four separate occasions over a 3 week period at I or -2 x
10'/rnL and counted
41

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
4-7 days later. No differences were observed in the t rowth rates of the cells
transduced with the
different constructs (Figure 8).
]00109] The results of these experiments show that both the shR A targeting
CCR5 and the 0446
protein can be expressed sufficiently fr-orn the same vector in human T cell
lines, and expression
of CCR5 shRNA and C46 has no effect on the growth rate of the cells.
Example 3. Transduction of Human Peripheral Blood Mononuclear Cells (PBMC) by
sta5/C46 Dual Vector
[00110] The various lentiviral vectors described in Example I were used to
infect human
peripheral blood mononuclear cells (PBMQ obtained from the Australian Red
Cross Blood
Transfusion Service, PBMC; were isolated from huffy coats using Ficoll-plaque
PLUS (GE
l-l_ealtheare) followed by CCD8 depletion using C ID H- Mierobeads (Miltenyi
Biotec) and a
VarioMACS magnetic unit. CD$-F depleted PBMC; were cultured for 48 hours in
RPMI 1640
media supplemented with 20% F13S and 5 g/inl, phy'to}her ragglutinin (PLIA)
(Sigma) at 2 x 106
cells/mL. Following 2 days PUr-\ stimulation, cells in suspension were
harvested, centrifuged at
200g for 5 mrrinutes and resuspended at 2x10`' cells%mL in RPMI -F- 20'/`0 F
13S -F- 10 LimL
recombinant human interleuhin-2 (rhlL-2; Roche) for 4 hours prior to
transduction.
100111] To ascertain the preferred transduction method, PBMC were transduced
with the
shy/EGFP lentiviral construct using various conditions: IX transduction with
WIM, 2X
transduction with WIM, IX transduction with VCM preload (Preload 1), 2X
transduction with
VCN'I preload (Preload 2), concentrated \JCM approximately 20-fold
(concentrated-)( 'see
Example 1, Section B). As shown in Figure 9, transduction was most efficient
with concentrated
virus. A single transduction with VG 'l preload (Preload. 1) was chosen as the
preferred method
for further experiments. MFI values for Figure 9 are shown in Table 5 below.
42

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
Table 5. Mean Fluorescence Intensity Values for PBMC expressing shS/ EGHI
construct
T ansdai
2x Transdn 1x Transdn 2x Transdn Cone VCM
VC1 VC14I
preload preload
---------------
------------- ---------
--------------------------------------- ---------------------------------------
----- -------------------- ----------------------------- ---------------------
----------------- -- --------------------
Replicate 1 25.9 20.0 22.5 16.0 16.8
eplic to 2 28.4
24.3 20.1 16.7 15.6
[00112] PBMC were either left untransduced or transduced (IX pre-load) with
one of shy/GFF,
C46, sh5/C46, GFP control or shy lentiviral constructs according to the
following procedure.
lrnL PBMC were transferred onto Retronectin coated 24-well plates (5 pg/crn)
previously pre-
loaded (6 hours) with 250 ,ul, ofunconcentratedVCM and cultured overnight. The
following
day, cells were transferred to 6-well plates in 3rnL. IIPM1 + 20% FBS
101.1/nnZ. rh1L-2. Cells
were analyzed for EC aFP, CCR5 and C46 expression 4 days post transduction.
The results are
shown in Figures 10-13.
1001131 As shown in Figure 10, expression ofEGFP, CCR5, and C46 in PBMC at day
4 post
transduction was as expected for the different constructs. EGFp expression was
observed for the
constructs containing EGFP (GFP control and shy/(iFE; panels 1,2); a reduction
in CCU
expression (illustrating expression of CCR5 shRNA) was seen for the constructs
containing shy
(sh5/EGFP and sh5/C46, panels 2 and 4), and C46 expression (as measured by 2F5
antibody)
was observed for the constructs containing C46 (C46 and sh5/C46; panels 3,4).
MF1 values, from
left to right in Figure 10, were 16.2, 8.4, 16.8, 9.4.
1001141 Figure 11 shows a comparison of gene expression in transduced 13BMC
(at day 4) and
transduced CEM.NKR.CCR5 T cell line (at week 8). EGFP expression was observed
in cells
transduced. with the constructs containing EGFP (GFP control and shy/GFP;
panels I and 2)1;
CCR5 down-regulation was seen in cells transduced with the constructs
containing; shy
(sh5/EGFP and. sh5/C46; panels 2. and 4), and C46 expression (as measured by
2F5 antibody)
was observed in cells transduced with the constructs containing C46 (C46 and
sh5/C46, panels 3
and 4). Although sufficient levels of expression from the lentiviral
constructs were observed in
4

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
both cell types, higher levels of expression were observed in the T cells as
compared to PBMC.
MFI values for Figure 11 are shown in Table 6 below.
Table 6. Mean Fluorescence Intensity Values for PBMC or CEM.N .CCRS T cells
expressing various constructs
GFP control Sh5/EGFP x:46 Sh5/C46
--------- --! ------------------------ ---------- ------------------------- ---
------------------------------------ ---------------------------------------¾--
-------------------------------------
I B IC 16.2 8,4 16.8 9.4
T Cell Line 153.6 18.6 129.1 22.6
----------------------------------------
[00115] In addition, growth rates were compared between gene-transduced (shy,
shy/EC aFP,
C46, sh5/C46) and non-transduced PBMC at days 1, 4, 8 and 12, Two replicate
seeds of each
group were used. The total cells/well and percentage ofviable cells were
similar for all
transduced P11MC when compared to one another and to the untransduced cells
(Figure 12).
[00116] Stability of transgene expression in PBMC' was also tested. Figure 1
shows expression
of EGFP, CCRS, and C46 (as measured by 2F5 antibody) in cells transduced with
the indicated
consfi-acts at days 4, 7 and 12. Viability of cells at day 12 was uncertain
and therefore
comparisons were made between days 4 and 7 only. As shown in Figure 1 3, the
various
transgenes were expressed at both time points with an apparent decline over
time, which is
probably related to decreasing growth and viability over time (see Figure 12).
MF1 values for
Figure 13 are shown in Table 7 below.
'Table 7. Mean Fluorescence Intensity Values for PBl 1C expressing various
constructs
CFP control S15 Shy/ECFP C46 Sh5/C46
Day 4 16.2 10.8 804 16.8 9.4
Day 7 13.2 12.6 8,7 14.4 10.8
[00117] These results show that both the shRNA targeting CCR5 and the C46
protein can be
11
expressed sufficiently from the same vector in human PBMC.
44

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
Example 4. ' 'raps uctlon of Human ematopoictic Progenitor/Stem Cells (HPSC)
by
sh5/C46 Dual Vector
[00118] The sh5/C46 lentiviral vector (L ~) was used to transduce CD34+
hematopoietic
progenitor/stern cells (HPSC) obtained from bulk donor peripheral blood
mononuclear cells.
Donors were injected with granulocyte colony-stipulating factor (G-CSF) to
mobilize HPSC and
peripheral blood mononuclear cells. Following G-CSF injection, the cells were
harvested by
apheresis and the bulk mononuclear cell population containing mobilized HPSC
were frozen,
The mononuclear cell sample used in this example was obtained from these
frozen stocks. A 50
ml, stern cell harvest bag estimated at the time of freezing to contain 3.7 x
107 CD34+ HPSC,
was thawed. On thawing, it was found to contain a total of 33.6 x 108 viable
cells (73% viability)
and the resulting number of CD34+ HPSC isolated using MACS (Magnetic Antibody
Cell
Separation) was within expectations at " 3.3 x 101 i.e. -1% of the total
mononuclear cell number,
with 98 Q CH34 positive (See pre- and post-separation analysis in upper panel
of Figure 14).
[00119] These cells were then used in the following experimental protocol:
1. 6 x l ()6 cells were pre-stimulated for 2.4 hours in X-vivo serum free
media
containing Stem Cell Factor (SCF), tlnronrbolzoietin (TPO) and F1t3 ligand
(Fit3I,) (each
at 50 ng/inL).
2. Aliquots of 4 x 105 cells were then transferred to a 12-well plate
preloaded for 6
hours with Virus Containing Medium (VCM) . The cells were transduced overnight
(with
GFP control, shy, shy/EGFP, C46, or sh5/C46), or left untransduced, and then
transferred
to fresh media for 72 hours.
3. FACS analysis performed 72 hours post-transduction demonstrated 25-30%
transduction by GFP (Figure 14, bottom panel). In this example, C46 was
undetectable by
2F5 staining apparently due to the lack of sensitivity of the flow cytometry
with these
cells. The remaining cells were put into CAMEO-4 (Hemogenix) metbyicellulose
cultures, plated at 1 00 cells/ well in replicate. When scored for colonies,
no significant
differences were seen between the cultures at day 11 (Table 8).

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
Table 8. Colony Percentages of CD34+ HPSC TransÃluceÃ1 with Various Lentiviral
Constructs
(_TU-E BF1-i-
CF - GM
E
Control 30.5 9.9 59.6
(untransduced)
(il/P control 15.6 28.9 55.6
---------------------------------------------------------- --------------------
-------------------------------------- ----------------------------------------
------------------- -----------------------------------------------------------
CFP control 18.5 9.3 72.2
sh5 41.2 11.8 47.1
---------------------------------------------------------- --------------------
--------------------------------------- ---------------------------------------
-------------------- ----------------------------------------------------------
--
sh5/Ef FP 2 a.0 30,0 44.0
shy/E(F'P 10.4 4.5 85.1
---------------------------------------------------------- --------------------
--------------------------------------- ---------------------------------------
--------------------- ---------------------------------------------------------
--
C46 10.2 28.6 61.2
C46 21.4 17.9 60.7
sh5i C46 17.6 35.3 47.1
sh5/C46 15.4 38.5 46.2
-----------------------------
Average 2t}.7 21.5 57.9
Std Dcv. 9.6 12.2 13.1
Example 5. sh5/C46 Dual Vector-Transduced T Cell Lines Inhibit HIV Replication
1001201 T cell lines (Mo1t4/CCR5) trarnsduced with the sl15/C46 dual
lerrtiviral construct (see
description of vector in Example 1) were challenged with various strains of
III : I-1IVBai ((-;CR5
tropic), IV-r1B (CXCR4 tropic), and IIVsF2 CC R5 and CXCR4 tropic). For
challenge assays,
1 x 1()6 transduced Molt4/CCR5 cells were added to 15 niL; tubes and
centrifuged. The
supernatant was discarded. 1-1 V virus containing medium (V CM) was added to a
filial
concentration per tube at a multiplicity of infection (MOD) of 11.2,-0.002.
Polybrene was then
added to a final concentration of 8 ~1g/niL and each tube was tapped gently.
Cells and virus were
incubated for 2 hours at 37 C with gentle shaking every 311 minutes. Following
the 2 hour
incubation, cells were washed in media (RPMI + 10% FBS) and resuspended in 34
mL of media
in T25 flasks. Cells were sampled and fed every 3-4 days until day It. 150 lul
of supernatant
was removed in duplicate and stored at 4 C. P24 protein levels (a measure of
IV infection) was
assayed as per Manufacturers' protocol generally -using 1/1()} to 1 /106
dilution to ensure values
were on Standard Curve.
46

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[001211 Figure 15 shows the p24 protein levels from non-transduced cells or
cells transduced
with the dual sh5/C46 lentiviral construct 13 days following challenge with
dual tropic HIV
strain SF2 (CCR5 and. CXCR4 tropic). The results show that cells transduced
with the sh5/C46
construct exhibited an approximate 2 log inhibition at all three MOls (O,2.
0.02, 0.002) in each of
2 independent samplings as compared to non-transduced cells. Figure 16 shows
the p24 protein
levels from non-transduced cells or cells transduced with either the sh5iC46
or C46 lentiviral
construct 11 days following challenge with dual tropic HIV strain SF2. The
data show
approximately 2 log inhibition by sh5/C46 construct in each of two independent
samplings and 3
log inhibition by C46 (apparently due to higher expression of C46 in this
particular construct) at
the two MOls tested. The bottom panel of Figure 16 shows expression by flow
cytometry. Mean
Fluorescence Intensity values are shown in Table 9 below.
Table 9. Mean Fluorescence Intensity Values for Molt4/CCR5 cells expressing
C46 or
sh5/C46lentiviral constructs
--------------------------------------- ---------------------------------------
-- --------------------------------------- ------------------------------------
---
Untr ansduced C46 Sh5/C46
CCR5 MIFI 1478.8 1811.0 44409
------------- ------------------------- ---------------------------------------
--- --------------------------------------- -----------------------------------
----
2FS MFI 344.6 1454,4 1171,8
1001221 In a separate experiment, lMolt4/CCR5 cells were either non-transduced
or transduced
with C'46 (Gene 2) or sh5/C46 6 (12R5) lentiviral constructs and subsequently
challenged with
HIV-SF/dual tropic CC R5 and (,XCR4), Bal (CCR5 tropic) or NL4-3 (("XCR4
tropic) virus at
an MOI of 0.2. P24 protein levels were assessed 11 days after viral challenge
as a measure of
HIV infection. As shown in Figure 17, cells expressing both lentiviral
constructs were effective
in reducing infection with all three strains of HIV. Figure 18 shows p2.4
protein levels from non_
transduced. cells (Moit4) or cells transduced. with one of four lentiviral
constructs [(I) shy (R5);
(2) C46 (G2); (3) sh5/C46 (R5-C2.); (4) shy./FCFP (R5-GFP) and 10 days
following challenge
with CCR5 tropic HIV strain Bal at a MOi of 0.2. The "mix" group is a mixture
of
untransduced, shy, C46, sh5/C46 all mixed equally (i.e. 25% of each). The
results show that
cells expressing the shR A against CCR5 and the C46 gene from a single
lentiviral construct
47

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
(dual construct) provide enhanced protection against infection with a CCR5
tropic HIV strain at
both" and 10 days following viral challenge.
[00123] The results of this series of experiments show that T cells transduced
with the dual
sh5/C46 lentiviral construct provide protection against infection with CCR5,
CXCR4, and dual
tropic CCR5 and CXCR4 HIV strains.
Example 6. shS/C46 Dual Vector-Transduced PBMC Inhibit HIV Replication
[00124] Phytohemagglutinin (PHA)/1L2-stimulated peripheral blood mononuclear
cells (P MC;)
were transduced with lentiviral vectors as described in Example 3, A schematic
of the dual
construct expressing shRNA against CCR5 and C46 protein (LVsh5C46) is shown in
Figure
Z--
1 9A. Four days post-transduction, cells were stained with the appropriate
monoclonal antibody
(e.g., CD195 or 2F5 antibody) and CCR5, CC46, and GFP expression was analyzed
by flow
cytametry (Figure 1911). Lentiviral (LN%)_ transduced PBMC~ were challenged
with R5 or X4
tropic HIV strains 16 days after 1-,V transduction. Culture supernatants were
collected four days
after HIV infection and assayed for p24 protein by ELISA (Figure 19C).
10}125] As shown in Figure 19C, P1BMCC transduced with sh5/C46 lentiviral
vector exhibit
reduced HIS' infection induced by both R5 and X4 tropic strains as assessed by
p24 protein
levels. PBMC, transduced with a shy/GFP construct are resistant to infection
induced by 1R5 but
not X4 tropic 1IIY. These results indicate that the sh5/C46 dual vector is
capable of protecting
against 1-11V infection induced by either R5 or X4 tropic strains.
Example 7. shy Vector Down Regulates CCR5 Expression in Lymphoid Organs and
Effects
Preferential Survival of Transduced C D4+ T Lymphocytes Ex Vivo
[00126] shy lentiviral-transduced C1)34-1 hei atopoietic progenitor/stet cells
(HPSC) solidified
with Matrigel in combination with a thymus segment were implanted under the
kidney capsule of
a humanized bone marrow/liver/thymus (BLT) mouse model (see Me us et a7.
(2006) .Nat Mcd,
Vol. 12:1316-1322; Shimizu et a7. (2010) .Blood, Vol. 115:1534-1544). The
NOD/SLID-hu BLT
humanized mouse allows examination of the differentiation of transduced human
HPSC in the
human thytruis-life organoid (thydiv), and migration of differentiated human T
lymphocytes in
48

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
systemic lymphoid organs including gut associated lymphoid tissue - the major
site of HIV
replication.
[001271 To examine sh1005 (shR`~l A targeting CCR5) in this humanized mouse
model, vector-
transduced fetal liver-derived CD 34+ cells and CD34- cells solidified with
matrisgrel and a
thymus segment were transplanted under the kidney capsule to generate a vector-
transduced
thy/liv tissue, Three weeks later, vector-transduced autologous CD34+ PIPSC (I
x_ 106 cells)
were injected through the tail vein of the sub-lethally irradiated mouse. To
assess the impact of
CCRS reduction within an animal, an equal mix of sh1005 vector (f GfP+)- and
non-shRNA
control vector (mdherry+)-transduced C"1 34+ HPSC (5 x 10 cells) were co-
transplanted, This
experimental design allows one to examine whether shl 005 vector-transduced
cells differ from
non-shRNA , vector-transduced cells with regard to level of stability and
specificity of CCR5
reduction; both vectors are present within the same animal to control for
mouse to mouse
variation. Neither ECipp nor mCherry alone had any effect on repopulation
kinetics or CCR5
expression (data not shown).
[0012811 uman cell engraftment was examined from 1 1 weeks post-C.D34-f
injection. Hurn -all
C, D45-j- lymphoid cells were detected in a gated lymphocyte population of
peripheral blood from
transplanted mice by flow cytometric analysis (average 441X"), SD---,-/- 8,
n=19). p,GFP and niCherm-yr
expression was found in this human C'D45-f population in transplanted mice
(average p.GFP
22%, SD 19, average mC;herry 22 %), SD+13, n-16). C'C'R5- ockdowvn in human
C,D4+ and
C, D45-+ 1-lymphocytes in various lymphoid tissues in reconstituted animals at
14-20 weeks post
CD34+ HPSC transplant was examined (Figure 20A). CCR5 expression was
efficiently reduced
in EGFP+ human C;D4+ and. CD45+ T-lymphocytes in all tissues analyzed.
Notably, CCR5
reduction was efficient even in the highly CCR5-expressing lamina propria
lymphocytes isolated
from the gut. CCR5 was not reduced in mCherr-y+ human CD4+/C'< 45+ T-
lymphocytes in the
same animal. These results indicate that the CCR5-shlUNA expression did not
affect human '-
lymphocyte differentiation and. migration and effectively induced CCR5 down-
regulation in
systemic lymphoid organs in vivo.
49

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[00129] To examine HIV susceptibility in CCR5 down-regulated cells. EGI P+ and
mCherry+
splenocytes were isolated from the animal by cell sorting. The sorted cells
were infected with
either R5 tropic IIIV-INFNSXS1.9 or X4 tropic HIV-I;NI.4-3 at a multiplicity
of infection of 2.5 in
triplicate. There was no increase in p24 HIV gate capsid protein production in
the culture
supernatant of EGF P+ splenocytes over the l 2 day culture period (Figure 20I3
). In contrast,
rnCherry+ splenocytes were susceptible to R5 tropic HIV-I NFNSXsL9, and
produced
approximately 4-fold higher levels of p24 in the culture supernatant on days 7
and 12 (P
value0 003), indicating CC R5 downregI lation effectively inhibited H5 tropic
HJV-1 infection.
In contrast to the H5 tropic HIV-1 infection, X4 tropic HIV-INI4-~ infection
produced
comparable amounts of p24 in both Ef 3FP+ and mC;hery+ splenocyte culture
supernatants
confirming the specificity of the inhibition (P value=0 ,23). These results
demonstrated that
downregulation of CCR5 by sh1005 was sufficient to protect ex vivo stimulated
cells from R5
but not X4 tropic 1-HV-1 challenge.
[00130] To examine 1-11V susceptibility in evo and selective protection and
survival of CCR5
dawn-regulated CD4 T-cells, R5 tropic HIV-1 NFNSxsrs was injected
intravenously (p24==200
ng) into reconstituted mice at 9 weeks post 1IPS(-' transplant. Mice were
confirmed to be infected
with HIV by the presence of p24 in supernatant of mouse peripheral blood co-
cultured with
111-IA!" 1L2 activated human PBMC at 8 week post IIIV injection. The kinetics
of EGFP and
mCherry expressing CD4 - T-cells in peripheral blood were examined Figure
20C:). Percent
EGPP-f- population in Cl)4+ T-cells in peripheral blood increased from 20% to
40'X") by 8 week
post HIV injection in the animals. In contrast, the mC'herry-- population in
CD4-1- -cells
declined from 40''0 to 3%% in the animals. The inversion of CD4/CD8 ratio
indicating HIV
induced CD4 'I-cell loss was examined. The CD4/CD8 ratio in EGF'P+ lymphocytes
was
maintained above I by 8 week post HIV challenge in peripheral blood (Figure
20D). In contrast,
CD4/CD8 ratio in mCherr-y+ CI)45+ cells was inverted to 0.1. These results
demonstrate stable
CCR5 down-regulation by shl005 was sufficient to preferentially increase CD4+
T-cells
following R5-tropic HIV challenge in vivo.

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
Example 8. 'Testing of sh5/C46 Dual Vector in Humanized Mouse Model
[001311 As described in Example 7 for the shy vector (Shimizu et al. (2010)
Blood, Vol. 115:
1534-1544). the sh5/C46 dual lentiviral vector is tested in the humanized BLT
mouse model. To
examine the sh5/C46 dual vector in this humanized mouse model. vector-
tranisduced fetal liver-
derived CD34+ cells and CD34- cells solidified with mnatrigel and a thymus
segment are
transplanted under the kidney capsule to generate a vector-transduced thy/hv
tissue. Three weeks
later, vector-transduced autologous CD34- 1=HPSC (1 x 106 cells) are injected
through the tail
vein of the sub-lethally irradiated mouse. To assess the impact of CCR5
reduction and C46
expression within an animal, an equal mix of the dual sh5/C46 vector (E FP+)-
and control
(en ty lentiviral) vector (mChe +)_transduced CT 34+ HPSC (5 x 105 cells) are
co-
transplanted. Other controls, such as shy single vector containing another
fluorescent protein
(e.g. YFP) and C46 single vector containing yet another fluorescent protein
(e.g. ('FP) are used
to transduce CD34-f- HPSC and are present in the mixture for transplantation.
This experimental
design allows one to examine the difference between cells transduced with
various constructs
with regard to level of stability and specificity of CCRS reduction and C46
expression-, all
vectors are present within the same animal to control for mouse to mouse
variation.
1001321 Control (m(.herry-) and active sh5/C46 fl_F 3FF-4,-) transduced cells
are compared over
time using flow cytometr and 1 1'rl'C1R. Comparisons are made between shy/C46-
transduced
cells and cells transduced with one of the single vectors (shy or ('146).
Susceptibility to HIV
infection is examined by injecting an R5, X4, or dual tropic 1-11V strain
intravenously into
reconstituted animals following 1-113SC transplant. Percent of CD4+ T-cells
and ratios of
CD4/CD8 T-cells in each of the vector-transduced populations is assessed to
ascertain the
effectiveness of CCR5 knockdown and C46 expression on CD4+ T cell survival.
Example 9. Use of Dual Constructs in Human HIV Patients
[001331 A dual lentiviral construct, including the sh5/C46 dual vector, the
sh5/TRIM5u dual
vector. or the shy/TRIM5a-cyclophilin dual vector, is introduced into
autologous human cells
and. subsequently provided to the patient. The dual lentiviral construct is
introduced into one or
more of CD34+ HP'SC cells, CD4+ 'I'-cells, CD8+ T-cells, monocyte/macrophages
isolated from
51

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
the patient to whom they will be re-implanted (e. g. autologous cells).
Alternatively cells from
another individual (allogeneic) are used. Alternatively a triple vector as
described herein is used.
[00134] With respect to HIV tropism, many patients will have R5 virus, a
smaller proportion will
have X4 virus, and an intermediate number will have a mixed population. The
dual constructs
described herein have the ability to target both R5 and r4 virus and can be
beneficial for patients
with a mixed cell population and may also prevent resistance in those with a
single population.
The constructs can also be beneficial in patients with HAART resistant virus.
[00135] The cells for transduction are obtained from the patient by injecting
one or more
cytokines that mobilize i-TPSC and other cells, and the relevant cell
popiilations are separated for
lentiviral transduction. The transduced-cells are intravenously introduced
into the same patient or
another patient in order to treat or prevent IITV infection. One or more doses
or infusions of the
transduced cells are used as described herein.
1001361 The clinical trial is designed based on considerations including the
patient's clinical
condition, previous treatment and/or resistance to treatment. Different
patient groups are
included in the trial. For example, one subset of patients has not yet
received highly active
antiretroviral therapy (i.e. naive to HAA RT). In general, these patients are
quite healthy
(notwithstanding their background HIV infection) and selection criteria for
receiving dual
lentiviral-transduced henratopoictic cells may include those patients who have
a history of a
relatively rapid CD4 decline, high viral load, and/or early symptoms. Figure
21 shows an
expected response in such a patient group. Dual lentiviral vector-transduced
cells are introduced
to a patient at time 0. Figures-2 IA and 21B show predictions of viral load
and CD4 count in
patients treated. with one infusion of transduced cells (star) versus patients
not receiving dual
lentiviral vector-transduced cells (triangle). The untreated. patients are
expected to maintain a
high viral load and a continuing decrease in CD4 count over time. In contrast,
those treated with
the dual lentiviral vector-transduced cells are expected to show viral load
decrease over time and.
CD4 count increase (after a potential initial small drop due to apheresis).
'Thus the treatment may
delay the need for H_<"ART and/or decrease its requirement once HAAF T is
initiated.
52

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
[00137] A second subset of patients is 1IIV positive and currently well-
controlled on III ART.
Figure 22 details an expected response to a single infusion of dual lentiviral
vector-transduced
cells in such a patient group, Predicted viral load for a patient treated with
one dose of
transduced cells (star) versus an untreated patient (triangle) is shown. Dual
lentiviral-vector--
transduced cells are introduced to a patient at time 0. Two HAART treatment
interruptions are
undertaken at various time points (ATI), e.g. from weeks 24-28 and 40-48, with
the patients
staying off IIA_ART if viral load remains below a pre-set safety limit (e.g.,
lOOK copies/rnL).
The HART interruptions are to provide a period where there can be HIV-induced
preferential
survival of those cells protected by the dual lentiviral construct and a
resulting decrease in viral
load. Primary end-point is at week 48 but one can also measure area under the
viral load curve
from weeks 40-48 and 40-100, Predicted viral load decreases in the long-term
for both treated
and untreated patients (though more slowly for patients not receiving dual
lentiviral-transduced
cell infusions) as the patients go back on HA ART (as required). The treatment
may decrease the
need for 1-1 AA RT and its associated complications.
1001381 A third patient group comprises individuals who are failing HAAR-T
because of
resistance to 1IAART drugs, non-compliance, or some other reason. Figure 2.3
depicts predicted
viral load (Figure 23)A) and predicted (_'D4 count (Figure 23113) expected in
such a patient. After
infusion of dual lentiviral-transduced cells at day 0, viral load is expected
to decrease and CD4
count to increase (star) as compared to an untreated patient where viral load
is expected to
remain the same or increase and CD4 count is expected to decrease with time
(triangle).
[00139] Endpoints in all of the patient groups include viral load, CD4 counts,
time to
resumption/Initiation of H '4 ART', transduced. cell percentage, and I-Cell
Receptor Excision
11 1
Circles (measure of recent thymic emigrants) and decreased requirement for
HST,
Example 10. Construction of Dual Vector Containing shRNA against CCR5 and
TRIMSa
(sh5I T M5a Dual Vector)
[00140] A dual lentiviral vector containing an shRN A targeting CCR5 under the
control of a HI
promoter and a nucleic acid encoding a TRTM5u protein under the control of a
ubiquitin
promoter is constructed using the backbone vectors described in Example 1. For
example, the U_

CA 02767972 2012-01-12
WO 2011/008348 PCT/US2010/036247
EGFP cassette is removed from pFG12-HI-R5-U-EGFP, the plasmid construct
containing
shI005 and ubiquitin promoter-driven EGFP (see Figure 3), using restriction
enzymes to produce
pFG 12-III -R5.
] The EGFP gene is removed from pFG 11 F -i_ -EGFP (pFG I I F in Figure 4) and
is 11 replaced with the TRI 5u. gene (SEQ ID NO: 5) to produce pFG 11F-U-
TRIM5a. The III-R5
cassette is excised from pFG12-Hi -R5-U-EGFP using an Mel / Xhol digest and is
inserted into
pFGI IF-U- TRIMSa, which has also been digested. with Ndel / Xhol, to produce
pFG1 IF-H1-
R5-U- TRIM5c. (Figure 24A). This construct is used to make lentivirus as
described in Section
B of Example 1.
Example 1.1. Construction of Triple Vector Containing shRNA against CCR5, C46,
and
T IM5Ãa (sti5/ C46/ T JM5aa Triple Vector)
1001421 A triple vector is produced from the dual vector pFG1 IF-1-I1-R5-U-
x:46 by cloning
11 ---actin promoter-TRIM5ct into a multicloning site as shown in Figure 24B.
[004431 All publications, patents and patent applications discussed and cited
herein are
incorporated herein by reference in their entireties. It is understood that
the disclosed invention is
not limited to the particular methodology, protocols and materials described
as these can vary. It
is also understood that the terminology used herein is for the purposes of
describing particular
embodiments only and is not intended to limit the scope of the present
invention which will be
limited only by the appended claims,
[001441 Those skilled in the art will recognize, or be able to ascertain using
no more than routine
experimentation, many equivalents to the specific embodiments of the invention
described
herein. Such equivalents are intended to be encompassed by the following
claims.
54

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Application Not Reinstated by Deadline 2023-07-11
Inactive: Dead - Final fee not paid 2023-07-11
Letter Sent 2023-05-26
Deemed Abandoned - Failure to Respond to Maintenance Fee Notice 2022-11-28
Deemed Abandoned - Conditions for Grant Determined Not Compliant 2022-07-11
Letter Sent 2022-05-26
Notice of Allowance is Issued 2022-03-11
Letter Sent 2022-03-11
4 2022-03-11
Notice of Allowance is Issued 2022-03-11
Inactive: QS passed 2021-12-10
Inactive: Approved for allowance (AFA) 2021-12-10
Examiner's Interview 2021-11-15
Amendment Received - Voluntary Amendment 2021-11-12
Amendment Received - Voluntary Amendment 2021-11-12
Inactive: Ack. of Reinst. (Due Care Not Required): Corr. Sent 2021-04-21
Reinstatement Request Received 2021-03-30
Amendment Received - Response to Examiner's Requisition 2021-03-30
Reinstatement Requirements Deemed Compliant for All Abandonment Reasons 2021-03-30
Amendment Received - Voluntary Amendment 2021-03-30
Deemed Abandoned - Failure to Respond to an Examiner's Requisition 2021-03-10
Letter Sent 2020-12-02
Inactive: Single transfer 2020-11-18
Examiner's Report 2020-11-10
Common Representative Appointed 2020-11-08
Inactive: Report - No QC 2020-10-23
Amendment Received - Voluntary Amendment 2020-02-18
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Inactive: S.30(2) Rules - Examiner requisition 2019-08-19
Inactive: Report - No QC 2019-08-05
Maintenance Request Received 2019-05-08
Letter Sent 2019-01-09
Letter Sent 2019-01-09
Inactive: Office letter 2019-01-08
Amendment Received - Voluntary Amendment 2018-12-20
Inactive: Reply to s.37 Rules - PCT 2018-12-19
Correct Applicant Request Received 2018-12-19
Inactive: Single transfer 2018-12-19
Inactive: S.30(2) Rules - Examiner requisition 2018-06-28
Inactive: Report - QC passed 2018-06-27
Maintenance Request Received 2018-05-10
Amendment Received - Voluntary Amendment 2018-01-03
Inactive: S.30(2) Rules - Examiner requisition 2017-07-05
Inactive: Report - QC failed - Minor 2017-06-30
Amendment Received - Voluntary Amendment 2016-11-04
Inactive: S.30(2) Rules - Examiner requisition 2016-05-04
Inactive: Report - QC passed 2016-05-03
Inactive: IPC deactivated 2016-01-16
Inactive: IPC removed 2015-09-22
Inactive: IPC assigned 2015-09-22
Inactive: IPC assigned 2015-09-22
Inactive: IPC assigned 2015-09-22
Inactive: IPC assigned 2015-09-17
Inactive: IPC removed 2015-09-17
Inactive: IPC removed 2015-09-17
Inactive: IPC assigned 2015-09-17
Inactive: IPC assigned 2015-09-17
Inactive: IPC assigned 2015-09-17
Inactive: IPC removed 2015-09-17
Inactive: First IPC assigned 2015-09-17
Inactive: IPC removed 2015-09-17
Inactive: IPC assigned 2015-09-17
Inactive: IPC assigned 2015-09-17
Inactive: IPC assigned 2015-09-17
Letter Sent 2015-05-14
All Requirements for Examination Determined Compliant 2015-05-01
Request for Examination Requirements Determined Compliant 2015-05-01
Request for Examination Received 2015-05-01
Change of Address or Method of Correspondence Request Received 2015-02-17
Inactive: IPC expired 2015-01-01
Maintenance Request Received 2013-05-24
Inactive: Cover page published 2012-03-16
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: First IPC assigned 2012-02-27
Application Received - PCT 2012-02-27
Inactive: Notice - National entry - No RFE 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: IPC assigned 2012-02-27
Inactive: Correspondence - PCT 2012-02-14
National Entry Requirements Determined Compliant 2012-01-12
BSL Verified - No Defects 2012-01-12
Inactive: Sequence listing - Received 2012-01-12
Application Published (Open to Public Inspection) 2011-01-20

Abandonment History

Abandonment Date Reason Reinstatement Date
2022-11-28
2022-07-11
2021-03-30
2021-03-10

Maintenance Fee

The last payment was received on 2021-04-22

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
THE REGENTS OF THE UNIVERSITY OF CALIFORNIA
CSL BEHRING GENE THERAPY, INC.
Past Owners on Record
DONG SUNG AN
GEOFFREY P. SYMONDS
IRVIN CHEN
LOUIS RANDALL BRETON
MAUREEN BOYD
MICHELLE MILLINGTON
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column (Temporarily unavailable). To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2012-01-11 54 4,217
Drawings 2012-01-11 24 1,593
Claims 2012-01-11 7 323
Abstract 2012-01-11 2 115
Representative drawing 2012-02-27 1 61
Cover Page 2012-03-15 2 99
Description 2012-01-12 60 4,353
Description 2016-11-03 60 3,616
Claims 2016-11-03 8 245
Description 2018-01-02 61 3,397
Claims 2018-01-02 8 239
Description 2018-12-19 61 3,404
Claims 2018-12-19 8 255
Claims 2020-02-17 8 250
Description 2021-03-29 55 3,278
Claims 2021-03-29 8 265
Claims 2021-11-11 8 265
Reminder of maintenance fee due 2012-02-26 1 111
Notice of National Entry 2012-02-26 1 193
Reminder - Request for Examination 2015-01-26 1 124
Acknowledgement of Request for Examination 2015-05-13 1 174
Courtesy - Certificate of registration (related document(s)) 2019-01-08 1 106
Courtesy - Certificate of registration (related document(s)) 2019-01-08 1 106
Courtesy - Certificate of Recordal (Change of Name) 2020-12-01 1 397
Courtesy - Acknowledgment of Reinstatement (Request for Examination (Due Care not Required)) 2021-04-20 1 404
Courtesy - Abandonment Letter (R86(2)) 2021-04-20 1 551
Commissioner's Notice - Application Found Allowable 2022-03-10 1 571
Commissioner's Notice - Maintenance Fee for a Patent Application Not Paid 2022-07-06 1 553
Courtesy - Abandonment Letter (NOA) 2022-09-05 1 546
Courtesy - Abandonment Letter (Maintenance Fee) 2023-01-08 1 551
Commissioner's Notice - Maintenance Fee for a Patent Application Not Paid 2023-07-06 1 550
PCT 2012-01-11 20 858
Correspondence 2012-02-13 2 76
Fees 2012-05-24 2 76
Fees 2013-05-23 2 75
Correspondence 2015-02-16 4 232
Examiner Requisition 2016-05-03 7 453
Amendment / response to report 2016-11-03 59 2,418
Examiner Requisition 2017-07-04 5 387
Amendment / response to report 2018-01-02 28 1,099
Maintenance fee payment 2018-05-09 1 60
Examiner Requisition 2018-06-27 6 374
Response to section 37 2018-12-18 4 127
Amendment / response to report 2018-12-19 28 1,072
Courtesy - Office Letter 2019-01-07 1 46
Maintenance fee payment 2019-05-07 1 59
Examiner Requisition 2019-08-18 3 187
Amendment / response to report 2020-02-17 12 398
Examiner requisition 2020-11-09 5 245
Reinstatement / Amendment / response to report 2021-03-29 26 1,050
Interview Record 2021-11-14 1 15
Amendment / response to report 2021-11-11 12 389

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :